Archives

Male Gonadal Disorders In The Tropics

ABSTRACT

 

Male hypogonadism arising from disorders of the hypothalamic-pituitary-gonadal axis is characterized by insufficient testosterone production. It is usually associated with subfertility or infertility. While hypogonadism is a global health concern, its diagnosis and management in tropical regions present unique challenges due to a combination of factors. Infectious etiologies often dominate the cause of male hypogonadism in certain areas of the tropics, but other factors such as environmental toxins, heat exposure, and high prevalence of metabolic disorders can also contribute. Atypical but not uncommon etiologies in the context of tropical conditions include snake envenomation, calorie deficiency, trauma, and androgen and recreational drug abuse. Understanding the specific causes of male hypogonadism in tropical regions requires a comprehensive assessment considering both medical and contextual factors. Addressing these causes involves targeted interventions, including infectious disease management, environmental regulations, genetic screening, appropriate medication use, and culturally sensitive healthcare approaches.

 

INTRODUCTION

 

Male gonadal function primarily refers to the role of the testes in producing testosterone and sperm. It is regulated by a complex interplay of hormones and feedback mechanisms. The hypothalamic-pituitary-gonadal (HPG) axis is the critical regulatory system that governs the function of the testes in producing sex hormones and sperm (1).

Male hypogonadism encompasses abnormalities in sperm production, including changes in quantity or quality, alongside androgen deficiency. In tropical regions, male hypogonadism can arise due to diverse factors such as heat exposure, nutritional deficiencies, infectious diseases, toxins, genetic disorders, and metabolic dysfunction. Effective management in tropical areas necessitates a comprehensive approach that takes into account environmental, nutritional, hormonal, and metabolic factors.

EPIDEMIOLOGY

 

The epidemiology of male hypogonadism remains insufficiently researched, particularly in tropical countries. Among the known causes of endogenous androgen deficiency, Klinefelter syndrome is relatively common, with a likely population prevalence ranging from 5 to 25 cases per 10,000 men (2). The percentage of infertile men varies widely, ranging from 2.5% to 12%. Infertility rates tend to be highest in Africa and Central/Eastern Europe (3).

In many tropical countries, endemic infections such as tuberculosis, leishmaniasis, leprosy, and schistosomiasis persist, leading to hypogonadism due to scrotal involvement (4). The precise prevalence, however, remains unknown.

INFECTIOUS CAUSES

Infectious causes of hypogonadism can result from various pathogens, including bacteria, viruses, and protozoa, that directly or indirectly affect the gonads or disrupt hormonal regulation. Bacterial infections ascending through the urogenital tract primarily affect the epididymis and accessory glands, whereas viral infections transmitted via the bloodstream predominantly involve the testes (5). Infections of the male genitourinary tract are responsible for 10% to 15% of cases of male infertility and may be especially relevant in the tropics (6). These conditions present as urethritis, prostatitis, orchitis, or epididymitis and are potentially curable (7).

The testis is considered an immune-privileged organ, crucial for safeguarding immunogenic germ cells during spermatogenesis from immune system activation. This protection is primarily achieved through a local immunosuppressive environment and systemic immune tolerance (8). The testis induces local innate immune responses to counter pathogens despite its immune privilege. However, certain pathogens can evade these defenses, leading to infection and persistence in the male reproductive tract (9).

Viral infections

 

Mumps virus and human immunodeficiency virus (HIV) infections are recognized viral causes of orchitis and male infertility. Additionally, various emerging viral infections, including tropical ones, can affect male gonads.

MUMPS

Mumps infection is known to cause hypogonadism and male infertility. The extensive use of mumps vaccines has reduced the occurrence and severity of mumps-related complications. In Asia, infection is more prevalent during summer months, and a correlation between increased temperature and humidity has been suggested (10). A possible cause of mumps outbreak in many tropical countries could be inadequate vaccine coverage.

Clinical orchitis is rare in prepubertal males but affects 15-25% of adult men about a week after parotitis. Infertility or subfertility occurs in about 30% of orchitis cases, likely due to germinal cell damage, ischemia, or immune responses to the infection (10,11). Germ cell failure is more common than androgen deficiency in mumps and related viral infections. Treatment during the acute phase is supportive, as no proven therapy prevents sperm cell damage. Universal vaccination remains the primary strategy for preventing mumps-related infertility (12).

HIV INFECTION

Epidemiology

Studies report low serum testosterone in HIV-positive men ranging from 13% to 40%, with a recent meta-analysis suggesting a 26% prevalence (13,14). Secondary hypogonadism accounts for up to 80% of the cases and is attributable to functional hypogonadotropic hypogonadism (FHH) (13). In tropical countries, socioeconomic factors such as poverty, limited education, and inadequate healthcare resources contribute to increased rates of HIV transmission and hinder access to testing and highly active antiretroviral therapy (HAART). Studies conducted in tropical Africa show a prevalence of hypogonadism ranging from 8.7% to 37% in men with HIV (15,16).

Etiology

 

HIV-specific factors, alongside traditional ones, contribute to testosterone deficiency in men with HIV. While some association exists between testosterone levels and HIV-related parameters, such as low CD4 count, uncontrolled HIV viremia, weight loss, and acquired immunodeficiency syndrome (AIDS) wasting, the evidence is not strong (17). The pathogenesis of hypogonadism in these men is multifactorial and complex, with classical risk factors playing a minor role compared to HIV-negative men. It's essential to note that the lack of a strong association between testosterone levels and traditional risk factors doesn't exclude their involvement; rather, numerous HIV-specific factors can mask their significance statistically (13).

HIV-related co-morbidities, chronic inflammation, illicit drug use, and body composition changes from HAART have been implicated in the development of hypogonadism. HIV infection makes the testes more susceptible to opportunistic infections like cytomegalovirus (CMV), Epstein-Barr virus, and tuberculosis (18). Up to 25% of individuals with AIDS will demonstrate testicular involvement with widespread opportunistic infection or systemic neoplasms, including CMV, toxoplasmosis, Kaposi sarcoma, and testicular lymphoma. However, primary hypogonadism may not develop in all cases(19).

Drug-Induced Hypogonadism

Several medications used for the treatment of HIV and AIDS may affect the HPG axis. Ketoconazole inhibits side-chain cleavage enzymes and other critical enzymes in testicular steroidogenesis. Megestrol acetate is used to increase appetite, but as a synthetic progesterone agent it suppresses gonadotropin secretion and results in hypogonadism. Central hypogonadism can also occur from opiate-induced inhibition of gonadotropin-releasing hormone (GnRH) release.

Hyperprolactinemia and Gynecomastia

Increased prolactin levels are reported in almost 20% of men living with HIV (20,21). In a case-control study, gynecomastia was seen in 1.8% of 2275 consecutively screened cases and was associated with hypogonadism, hepatitis C, and the degree of lipoatrophy associated with HAART (22). Efavirenz, a commonly used HAART, is often responsible for gynecomastia which is due to direct activation of the estrogen receptor (23). Hyperprolactinemia has been reported in 21% men with stable disease and was significantly associated with opioid and protease inhibitor usage.

Testicular Changes

HIV infection itself doesn't result in observable morphological changes, especially with the advent of HAART, which has majorly reduced the risk of primary testicular damage (24). An earlier autopsy-based study had categorized testicular findings in AIDS into five groups: "Sertoli cell-only" syndrome (43%), germ cell damage (27%), peritubular fibrosis (15%), maturation arrest (12%), and normal appearance (3%) (25). A subsequent study reported decreased spermatogenesis, subacute interstitial inflammation, or their combination in autopsy (26).

 

Diagnosis and Management

 

The approach to diagnosis and management is generally similar to other causes of male hypogonadism. Readers can refer to relevant sections in endotext.com for more detailed information (27–29). Of note, about 30% to 55% of men with HIV have increased sex hormone–binding globulin (SHBG). As a result, using bioavailable or free testosterone instead of total testosterone is recommended for diagnosis. Though, in cases of hypogonadotropic hypogonadism, addressing the primary pathology is the standard treatment, the chronic nature of the condition demands more frequent consideration for testosterone replacement therapy (TRT) for men with hypogonadism and HIV (30).

Treatment options include TRT, addressing underlying comorbidities, optimizing HAART regimens to minimize side effects, and promoting healthy lifestyle practices to prevent metabolic disorders. Regularly monitoring hormone levels, bone health, and metabolic parameters is crucial for long-term management.

ZIKA VIRUS INFECTION

Zika virus is a flavivirus borne by mosquito vectors such as Aedes aegypti and Aedes albopictus. It is endemic to tropical countries of Africa, Asia, and South America. The virus can also spread through sexual contact, blood transfusion, and from mother to fetus (31).

The infection remains asymptomatic in the majority, but manifestations may include low-grade fever, rash, conjunctivitis, myalgia, and arthralgia. Zika virus RNA persists in the semen and in male and female reproductive tracts. Zika virus has been associated with testicular inflammation and damage, leading to infertility in some cases (32,33). The virus's ability to alter mature sperm can reduce fertility and has implications for assisted reproduction, particularly due to its teratogenic potential (34). Typically, the testes do not show any inflammatory response, and normal morphology and hormone production are maintained. This enables the virus to remain dormant, acting as a covert carrier for asymptomatic sexual transmission.

OTHER VIRAL INFECTIONS

Several viruses prevalent in tropical countries have been linked to testicular damage and infertility. Human papillomavirus (HPV) infection in males is often linked to external genital warts, but asymptomatic infections are equally common. HPV has been detected in the epididymis, testicles, vas deferens, prostate, and seminal fluid. High-risk HPV strains such as HPV-16 can affect sperm parameters, including count and motility, possibly reducing fertility (35,36). Both herpes simplex virus (HSV)-1 and HSV-2, like HPV, can localize in the male genital tract, but it's unclear if they affect fertility (37).

Hepatitis B virus (HBV) can enter male germ cells by crossing the blood-testis barrier, integrating its genome, and inducing oxidative stress and reactive oxygen species (ROS) production, leading to sperm apoptosis. HBV infection in chronic cases results in higher apoptotic sperm cells and membrane integrity loss (38). Despite its effects on sperm, fertility outcomes in assisted reproduction remain unaffected, with vertical transmission being unlikely, especially with a vaccinated female partner (39).

Hypogonadism has been documented in men infected with the hepatitis C virus (HCV), but the etiology has not been clearly established and is likely to be multifactorial. While systemic inflammation associated with HCV may suppress the HPG axis, the effect of advanced liver disease on testosterone metabolism may also be responsible (40). HCV infection reduces sperm count, motility, and morphology, affecting fertility potential. Elevated oxidative stress can lead to sperm chromatin condensation and cell death. It can also trigger an autoimmune response. Interestingly, treatment with ribavirin and interferon can also worsen semen parameters (41).

Male reproductive organs have been found to be vulnerable in moderate to severe illness with severe acute respiratory syndrome coronavirus 2 (42,43). The negative effect on seminal parameters was found to persist even at six months (44).

 

Bacterial Infection

Bacterial infections in the male reproductive tract can lead to epididymitis, orchitis, prostatitis, and urethritis. These infections are typically caused by Chlamydia trachomatis, Neisseria gonorrhoeae, ureaplasmas, mycoplasmas, and other bacteria. They are more common in tropical developing countries. Mycobacterial affection of the male genital tract is also prevalent in these regions. Symptoms include pain and swelling of the genitalia, penile discharge, and discomfort during urination or ejaculation. Treatment usually involves antibiotics targeted at the specific bacteria causing the infection (45).

Infertility can result from these infections, with underlying mechanisms possibly including damage to the germinal epithelium, ischemia, immune dysfunction, and cell damage from increased ROS (46). Spermatozoa can be affected at various stages of their development, maturation, and transport. Infections are also associated with obstruction along the seminal tract, such as urethral strictures.

Many pathogens of the male genitourinary tract are asymptomatic, and it is often difficult to distinguish colonization from infection detrimental to fertility (47). Bacteriospermia is suspected when there are more than one million peroxidase-positive white blood cells per milliliter of ejaculate (leukocytospermia). It is confirmed through a semen culture or polymerase chain reaction (PCR) to identify the pathogen. Antibiotic treatment may improve sperm quality and prevent testicular damage and complications, but its effects on natural conception are not clear (48). Furthermore, leukocytospermia is a sign of inflammation and may not be associated with a bacterial or viral process, hence its clinical significance in the ejaculate is controversial (49).

CHLAMYDIA

C. trachomatis, an intracellular gram-negative bacterium, causes asymptomatic infection of the genital tract in 85%–90% of cases. Symptoms of epididymo-orchitis and prostatitis include mucoid or watery urethral discharge and dysuria. Some but not all studies have demonstrated an association with male infertility and altered semen quality (45,50,51).

While some research suggests that C. trachomatis could affect sperm-egg penetration, impacting fertilization potential, others propose that its impact on male fertility might be related to transfer to a female partner and resulting inflammatory processes, anti-sperm antibody generation, or defective implantation. Overall, the association between C. trachomatis and male fertility remains complex and may vary depending on individual cases (45).

NEISSERIA

N. gonorrhoeae is a leading cause of genital infection in the tropics. It primarily spreads through sexual contact and can lead to asymptomatic colonization or inflammatory diseases like urethritis, orchitis, prostatitis, and epididymitis. These infections can manifest as mucopurulent urethral discharge, or infertility from testicular damage or ductal obstruction. The bacteria attach to spermatozoa using pili or direct contact, and their infection triggers an influx of inflammatory cells. While the exact causative role of N. gonorrhoeae in pathogenesis of male infertility remains unclear, studies have noted higher infection rates in men with infertility compared to those without fertility issues (52).

GENITAL UREAPLASMAS AND MYCOPLASMAS

Of the genital ureaplasmas and mycoplasmas, Ureaplasma urealyticum, and Mycoplasma hominis are potentially pathogenic and can contribute to both genital infections and male infertility (53,54). The prevalence of U. urealyticumranges from 10 to 40%. Both U. urealyticum and M. hominis have been linked to prostatitis and epididymitis (45). The mechanism of infertility could be due to a reduction in ejaculate's oxidoreductive potential, making sperms more susceptible to peroxidative damage (55).

LEPROSY

Leprosy is a chronic infectious disease caused by Mycobacterium leprae, primarily affecting the skin, peripheral nerves, mucosa of the upper respiratory tract, and eyes. The condition is prevalent in tropical countries, and according to World Health Organization (WHO) estimates, over 17 million patients received multidrug therapy (MDT) for leprosy in the past four decades. The lower temperature of the scrotal contents, between 27–30˚C, makes the testes prone to infection in those with the lepromatous form and during flares of erythema nodosum leprosum (type 2 reaction).

The testes can serve as a reservoir for leprosy bacilli, potentially leading to testicular atrophy through the mediation of inflammatory cytokines and endarteritis, ultimately resulting in fibrosis. Early symptoms include testicular pain or swelling. Hypogonadism can lead to decreased or absent libido (28%), followed by gynecomastia (16.3%). Smaller, softer, and less sensitive testes is a characteristic feature of leprosy. Ultrasonography demonstrates reduced testicular volume in 72% of affected males (56). Laboratory investigations reveal oligospermia or azoospermia, elevated luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and low serum testosterone levels (57–59).

TUBERCULOSIS

Epidemiology

Male genital tuberculosis is found worldwide but is more common in regions with high tuberculosis prevalence, such as parts of Asia, Africa, and Latin America. Genitourinary involvement accounts for 20-40% of extrapulmonary forms.Isolated genital infection is uncommon and occurs in 5–30% of the cases of genitourinary infection (60). Clinical reports likely underestimate the actual prevalence of male genital tuberculosis as symptoms are often absent (61).

Mode of Infection

 

Male genital tuberculosis typically originates from bacillaemia following primary infection of the lungs. Older studies suggest that the prostrate is often seeded by infected urine, with subsequent canicular or lymphatic spread to the epididymis (62). Though current literature suggests that direct hematogenous spread may be the primary mode of initial genital infection, especially in miliary cases. Granulomas formed systematically during primary infection can harbor bacilli for long periods, and reactivation can lead to genital tuberculosis. Disease progression often involves adjacent sites through direct extension, with orchitis almost always occurring secondary to epididymal disease. Concurrent or sequential involvement of multiple genital sites is common (63).

Clinical Features

Epididymis and prostate are the most commonly affected sites. Epididymitis is the most frequently reported form of male genital tuberculosis, characterized by gradual onset of swelling and pain. Acute infections are also observed. Spread to the testis can manifest as non-tender testicular mass, with coexisting enlarged, hard epididymis, beaded vas deferens, and sometimes scrotal edema. Oligospermia or azoospermia can occur from occlusion or granulomatous destruction of vas deferens or epididymis. Prostatic tuberculosis may present with dysuria, frequency, hematuria, and hemospermia. Physical examination may reveal firm enlargement, nodularity, or soft areas of necrosis (61,63).

Diagnosis and Treatment

 

Diagnosing male genital tuberculosis often requires a combination of clinical evaluation, imaging studies (such as ultrasound or magnetic resonance imaging), laboratory tests (including semen analysis, urine analysis, and tuberculosis-specific tests like PCR or culture), and sometimes biopsy of affected tissues. All patients with genital tuberculosis should be screened for pulmonary and renal lesions. Treatment typically involves conventional tuberculosis chemotherapy courses. In cases of infertility or complications, additional management strategies such as surgical interventions or assisted reproductive techniques may be considered. Early recognition and treatment are crucial in managing male genital tuberculosis and preventing complications such as infertility (64).

Other Mechanisms of Gonadal Dysfunction

Central nervous system tuberculosis, including tuberculomas involving the sellar region, can lead to hypogonadotropic hypogonadism (65). Pro-inflammatory cytokines, such as tissue necrosis factor-α (TNFα), interferon-γ, and interleukin (IL)-6, have been implicated in the impaired production of gonadal androgens in cases with pulmonary tuberculosis. These cytokines can disrupt the normal functioning of Leydig cells, leading to reduced testosterone synthesis (66).

OTHER BACTERIAL INFECTIONS

Brucellar epididymo-orchitis is a rare infection affecting the testis and epididymis, occurring in approximately 2–14% of cases of brucellosis. Brucellosis is still prevalent in individuals dealing with livestock in developing countries and is reported to be hyper-endemic in Iran. Necrotizing orchitis, testicular abscess, infarction, atrophy, suppurative necrosis, azoospermia, and infertility can occur if diagnosis is delayed or management is inappropriate (67).

 

Several other bacteria, such as Escherichia coli, Staphylococcus aureus, Enterococcus faecalis, Streptococcus agalactiae, Gardnerella vaginalis, Treponema pallidum, Helicobacter pylori have been linked to male infertility through different mechanisms (45). However, more research is needed to fully comprehend their roles, particularly in tropical regions where these bacterial infections are more prevalent.

Protozoa

 

Protozoan parasitic diseases are endemic in many tropical countries. Protozoan infections of the male genital tract are rare, and only a few species, such as Trichomonas vaginalis, Trypanosoma species, Leishmania donovani, Entamoeba histolytica, Acanthamoeba, Toxoplasma gondii, and Plasmodium falciparum, have been linked to pathogenesis of testicular damage (68).

TRICHOMONAS

T. vaginalis is a common sexually transmitted infection that can affect various parts of the male genital tract, including the urethra, prostate, and epididymis. Although uncommon, T. vaginalis can impact male fertility. Studies indicate a higher prevalence of T. vaginalis in infertile men compared to fertile individuals, and its presence in semen is linked to decreased sperm motility, normal morphology, and viability. In vitro studies confirm that T. vaginalis and its secretions can reduce sperm motility and fertilizing capacity (68,69).

TOXOPLASMOSIS

Congenital toxoplasmosis is characterized by meningoencephalitis with significant perivascular inflammation, particularly in the basal ganglia and periventricular regions. This condition likely affects important hypothalamic regulatory centers, resulting in hypothalamo-pituitary dysfunction. The clinical features of toxoplasmosis stem from both direct tissue destruction by the parasite and immunopathological changes mediated by inflammatory cytokines. Hypothalamic-pituitary dysfunction, precocious puberty, and central diabetes insipidus with hypogonadism have all been described in association with congenital toxoplasmosis (70–73). In immunocompromised individuals, such as individuals with AIDS, the male reproductive tract can rarely be affected, leading to conditions like epididymitis or orchitis. Although direct links to infertility aren't fully established, some studies suggest potential negative impacts on sperm health.

LEISHMANIASIS

Infections with Leishmania can lead to genital lesions and testicular amyloidosis, contributing to hypogonadism. Parasitism of the testes and reduced testicular size with fewer Sertoli and Leydig cells have been reported (74). Evidence of involvement of several endocrine organs- pituitary, adrenal, thyroid, and sex glands- via histopathologic studies have been documented in Visceral Leishmaniasis (75). However, abnormal endocrine function tests in some instances without clinical manifestations have been documented. Genital leishmaniasis lesions on the penis, mimicking a painless, slow-growing scabies-like ulcers, can occur uncommonly (76).

TRYPANOSOMIASIS

African trypanosomiasis, also known as sleeping sickness, is caused by the protozoan parasite Trypanosoma brucei, which is transmitted by the bite of a tsetse fly. In a study involving 31 Congolese men with confirmed trypanosomiasis, 70% experienced impotence, and 50% exhibited decreased testosterone levels (77). The gonadotrophins were found to be disproportionately normal, suggesting hypothalamic-pituitary involvement (78). The endocrine dysfunction observed in patients with trypanosomiasis may be secondary to inflammatory cytokines (79,80). However, further studies are required to confirm the hypothesis.

Chagas disease, caused by Trypanosoma cruzi, affects 6–7 million people worldwide, mainly in Latin America. It is primarily transmitted by triatomine bugs, with congenital and blood-transfusion transmission also reported. In its chronic phase, the disease commonly leads to cardiac, digestive, or neurological disorders. Early antiparasitic treatment can cure the acute phase, while treatment during the chronic phase can slow progression (81). Animal experiments demonstrated the presence of amastigote forms in seminiferous tubules of infected mice (82). Subsequent autopsy studies only revealed focal chronic phlebitis and mononuclear interstitial infiltration of the testis and failed to show any parasites (83). Early studies of testicular biopsies in chronic Chagas disease revealed arrested germ cell maturation and regressive alterations, worsening progressively from normospermia to azoospermia (84). Immune neuro-endocrine disturbance could possibly play a role in the pathogenesis (85).

OTHER PROTOZOAL INFECTIONS

Rare cases of scrotal and penile amebiasis have been described (86,87). Rare reports in the medical literature have mentioned cases where infections caused by Plasmodium falciparum or Plasmodium vivax, the parasites responsible for malaria, have led to testicular pain or hypogonadism (88,89).

Fungus

 

CANDIDA

Fungal epididymitis, caused by Candida glabrata, is uncommon but should be considered, especially in individuals with diabetes and a history of catheterization or antibiotic use. Rare cases with enlarged and tender hemiscrotum responding to fluconazole and surgical excision have been described (90). The risk of epididymitis in individuals with diabetes with C. glabrata and C. albicans increases with urinary tract instrumentation and prior antibiotic therapy. Diagnosis relies on recognizing fungi in histology or pus cultures, often indicating retrograde spread from urine. Fungal epididymo-orchitis can occur as an isolated entity or, more often, during disseminated infection (91).

As with any gonadal infection, fungal epididymo-orchitis can cause infertility because of gonadal destruction and resultant azoospermia. In addition to invading tissue, fungi can contribute to infertility by inducing anti-sperm effects and secreting mycotoxin. C. guilliermondii and C. albicans are able to inhibit sperm viability and motility in vitro. A proportion of infertile men and women have antibodies positive for C. guilliermondii, the implications of which are unknown. Restoration of fertility was achieved in some patients after the eradication of C. guilliermondii by ketoconazole (92).  

 

OTHER FUNGAL INFECTIONS OF GONADS

Other fungi reported to infect testis and epididymis include blastomycosis, histoplasma, aspergillus, and cryptococcus (93–95). Cryptococcus neoformans can also cause hypospermia and teratospermia (96). The fusarium toxin zearalenone and its metabolite zearalenol bind as agonists to estrogen receptor-α and -β, causing hyperestrogenism-mediated decreases in testosterone and libido, azoospermia, and feminization in mammals. Whether such hyperestrogenic effects occur in humans with fusariosis is unclear (97).

Granulomatous epididymo-orchitis can also occur as a part of disseminated histoplasmosis in an immunocompromised state (94). Genital blastomycosis is described mostly as a part of disseminated disease. Majority present with unilateral or bilateral pain and swelling of the scrotum. Onset can be acute or insidious, with symptoms lasting from days to months. Bacterial infection on the other hand is typically unilateral and acute (93). Some fungal infections may remain asymptomatic and only get detected during autopsy.

PITUITARY FUNGAL INFECTIONS

Pituitary fungal infections or abscesses are extremely unusual and mostly found in immunocompromised states. (98). The mode of spread could be hematogenous, extension from adjacent structures like meninges, sphenoid sinus, cavernous sinus, and skull base, or iatrogenic during transsphenoidal procedures. Aspergillus is the most frequently reported fungal infection of the pituitary (99). Candida, Pneumocystis jirovecii in HIV/AIDS, and coccidia are also reported to infect the pituitary (100–102).  Gonadotrophin and other pituitary hormone secretion can be affected, but such reports are very rare (103). Pituitary stalk compression due to fungal lesion can induce hyperprolactinemia (104).

Helminths

 

SCHISTOSOMIASIS

Schistosomiasis, caused by Schistosoma haematobium, S mansoni, and S. japonicum, represent a major tropical disease transmitted through contact with infested freshwater. S. haematobium, common in sub-Saharan Africa, infects around 112 million people and often affects the urinary tract, with potential extension to the genitalia. The infection can persist for decades and, if untreated, becomes chronic, with potential for causing complications (105). S. manson, iprevalent in the Caribbean, South America, and Africa, and S. japonicum in Southeast Asia are primarily associated with hepato-intestinal infection with very rare occurrence of genital disease. Genital involvement is primarily observed with S. haematobium (106).

Early symptoms include hemospermia, that results from mucosal ulceration caused by egg penetration into the seminal vesicle. Schistosoma eggs can become entrapped in the prostate, vas deferens, epididymis, or testes, and trigger immune reactions and granuloma formation. Clinical features include genital or ejaculatory pain, infertility, and abnormally enlarged organs from granulomatous infiltration, fibrosis, and calcifications (105–107). Diagnosis depends on identifying ova in semen or urine, but detecting chronic infection is challenging as ova might often be absent. Praziquantel (at 40 mg/kg) is the standard treatment for most forms of schistosomiasis (106).

S. mansoni infection has been associated with low normal testosterone and elevated estrogen levels in males, although hepatic dysfunction may play a role in these abnormalities (108).

FILARIASIS

Filariasis is a neglected tropical disease transmitted by mosquitos caused by Wuchereria bancrofti, Brugia malayi, and B. timori. Filariasis occurs in Africa, Asia, South America, the Caribbean, and the Pacific. Globally, it is estimated that 25 million men have hydrocele due to lymphatic filariasis, and over 15 million people are affected by lymphoedema (109). Initial infections are often asymptomatic, but chronic disease can damage the lymphatics of the spermatic cord. Common genital manifestations include recurrent scrotal pain and swelling, hydrocele, and epididymo-orchitis (110). Azoospermia and oligospermia are also described (111). The WHO's Global Programme to Eliminate Lymphatic Filariasis (GPELF) was launched in 2000 with a strategy focused on large-scale annual treatment in endemic areas to stop infection spread and provide essential care.

Ecdysteroids are compounds related to 20-hydroxyecdysone, the insect molting hormone, in Loa Cystoids and Mansonella perstans infections, the other form of filariasis. Microfilaremic males with these infections had low testosterone in 12%, and high gonadotrophins in 24%, and abnormal levels of both in 21%. Ecdysteroids were found in the serum of 90% of individuals with microfilaremia and in all urine samples, but their levels did not correlate with hormonal changes. A possible link between microfilaremia and endocrine disruptions, including hypogonadism, has been suggested, but the direct role of parasitic ecdysteroids remains unproven (112).

 

ENVIRONMENTAL CAUSES

 

Endocrine Disrupting Chemicals (EDCs)

DEFINITION AND CONTEXT

EDCs pose a significant and ubiquitous threat to global and tropical health. EDCs include both natural and synthetic chemicals widely dispersed in the environment. These chemicals can be ingested, inhaled, or absorbed through various media, including food, water, air, and consumer products, and can interfere with any aspect of hormone action. EDCs can bind to hormone receptors, such as estrogen and steroid receptors, disrupting development and reproductive function, among many other health impacts.

Common EDCs include bisphenol A (BPA), found in plastics and food containers, and phthalates, used to make plastics more flexible and present in products like cosmetics and toys. Polychlorinated biphenyls (PCBs), industrial chemicals in electrical equipment and paints, and dioxins, by-products of industrial processes and combustion, are also significant EDCs. Pesticides such as dichlorodiphenyltrichloroethane (DDT) and glyphosate, widely used in agriculture represent another major group of EDCs. For more details, please refer to the sections on EDC in Endotext (113).

EDCS IN TROPICS

Despite growing recognition of their impact, the full extent of their damage remains inadequately addressed due to insufficient evidence and lack of comprehensive testing (114). In tropical regions, extensive use of pesticides and industrial chemicals increases exposure to EDCs. For example, glyphosate, a commonly used herbicide, has been linked to endocrine disruption and adverse reproductive health outcomes (115)​. Similarly, heavy metals like lead and arsenic, prevalent in some tropical areas, cause significant endocrine-related health issues ​(116,117).

A review of data on prioritized EDCs (e.g., DDT, lindane, PCBs, etc.) reported elevated concentrations in the Indian environment and human population compared to the international context (118). A recent nationwide pilot study has reported the widespread occurrence of per- and polyfluoroalkyl substances (PFASs) and phthalates in humans from different locations across India, including those residing along the Indian Himalayas (119,120). Both DDT and pyrethroids used for malaria control in African countries have endocrine-disrupting potential (121).

EDCS AND MALE GONADAL DYSFUNCTION

Hypogonadism

 

EDCs act as anti-androgens, mimic estrogens, and inhibit steroidogenic enzymes, interfering with androgen production and function. Phthalate esters like di-(2-ethylhexyl) phthalate (DEHP) and BPA can reduce testosterone synthesis and disrupt gene expression related to hormone balance. DDT, PCBs, and their metabolites can also block hormone receptors, affecting estrogen and androgen signaling crucial for spermatogenesis and testicular development (122).

 

Infertility

 

EDCs are known to disrupt hormonal balance and have been linked to impaired sperm production, quality, and function. Factors such as type of EDCs, duration of exposure, and individual susceptibility play roles in their effects on reproductive health. EDCs impact sperm function by targeting testicular development and influencing the HPG axis, affecting estrogen and androgen receptors, influencing ROS production, inducing epigenetic modifications, and directly affecting spermatozoa and testicular tissue cells (123). Pesticides have been extensively studied for their effects on sperm parameters and DNA integrity. While some studies report reductions in sperm concentration and alteration in sperm morphology due to pesticide exposure, others show no significant impact (124). DDT, BPA, and phthalates are associated with decreased semen volume and sperm concentration, motility, and abnormal morphology (125). Increased urinary BPA level is associated with reduced number, motility, and sperm vitality, leading to male infertility (126). Continued research is needed to better understand the effect of EDCs on reproductive health.

 

Developmental Disorders

 

Testicular dysgenesis syndrome (TDS) is a condition linking poor semen quality, testicular cancer, undescended testes, and hypospadias. Experimental and epidemiological studies indicate that TDS stems from disturbances in embryonic programming and gonadal development during fetal stages. These disorders share a common pathway by which environmental chemicals and genetics result in abnormal development of the fetal testis (127,128). Though harmful effects on testicular development in animals have been demonstrated, the current evidence does not conclusively clarify the impact of EDCs on human male reproductive development (129).

 

Gynecomastia

 

Gynecomastia prevalence has increased over recent decades, partly attributable to exposure to EDCs. Higher plasma concentrations of DEHP and its major metabolite mono(2-ethylhexyl) phthalate (MEHP) in boys with gynecomastia have been demonstrated (130). Another study reported an outbreak of gynecomastia linked to the anti-androgenic delousing agent phenothrin (131). Additionally, essential oils like lavender and tea tree oil have been associated with gynecomastia. Components of these oils have estrogen receptor (ER) agonist activities (132). Occupational exposure to gasoline vapors and combustion products may play a role in the causation of male breast cancer (133).

 

Current literature indicates a possible link between EDC exposure and development of gynecomastia. Increasing rates of the condition indicate that environmental factors are important to disease etiology. The data from tropical countries is sparse, and epidemiological studies to evaluate the influence of EDCs on diseases of the male reproductive tract, including gynecomastia, are necessary (134).

 

Testicular Cancers

 

Few studies have explored the correlation between EDC exposure and testicular cancer, and even less so in tropical countries. The results are inconsistent, with some but not all studies showing an association between pesticide exposure and testicular cancer. Dichlorodiphenyldichloroethylene (DDE), chlordane, and PCB exposure have been linked to testicular cancer (135–137). These mixed findings highlight the need for more focused research on EDCs and testicular cancer, especially in tropical countries with high exposure to pesticides (129).

PREVENTION

Reducing exposure to EDCs through lifestyle changes, environmental regulations, and occupational safety measures can help mitigate their potential impact on male gonadal disorders. Additionally, further research is needed to understand better the mechanisms by which EDCs affect male reproductive health and to develop strategies for prevention and treatment.

Temperature

 

Heat exposure is a significant factor in male infertility, affecting sperm production and quality. Global warming and episodes of heat stress, occupational exposure, and lifestyle factors can be responsible for increasing scrotal temperature (138).

The testes are located outside the body in the scrotum to maintain a temperature of 2-4°C below core body temperature, optimal for spermatogenesis. A recent meta-analysis concluded that high ambient temperatures in tropical climates can negatively affect sperm quality, including decreased semen volume, sperm count, sperm concentration, motility, and normal morphology (139). This may be especially relevant for men working in high-temperature environments (e.g., welders, bakers, and drivers) or exposed to prolonged heat (e.g., saunas and hot tubs) (140,141). Studies have shown that even temporary exposure to high temperatures can significantly impact sperm parameters (142).

Similarly, febrile illness, prolonged sitting during work or truck driving, tight-fitting underwear, and laptop use with increased heat to the testes have been proposed to affect male fertility adversely (146,147). Studies in men have shown that small increases in testicular temperature accelerate germ cell loss through apoptosis. The data to support these associations are, however, inconsistent (143).

 

Trauma

 

Traumatic injuries to the genitalia, common in tropical regions due to occupational hazards, accidents, and interpersonal violence, can cause direct damage to the testes. Severe trauma can result in testicular rupture or vascular compromise, leading to hypogonadism due to impaired blood supply or loss of testicular tissue. Radical prostatectomy or other overt genital tract trauma is a physical cause of a sudden loss of male sexual function (144).

Males who experience a traumatic pelvic fracture or genital trauma may also have psychogenic erectile dysfunction (145). Post-traumatic hypopituitarism is responsible for about 7.2% of all causes of hypopituitarism and can develop after road traffic accidents, sports injuries, blast injuries, and other trauma. Peripherally placed somatotrophs and gonadotrophs are first affected by ischemic damage, while centrally located corticotrophs and thyrotrophs are subsequently involved (146).

 

Snake Envenomation

Snakebite envenoming is a medical emergency prevalent in tropical regions of Asia, Africa, and Latin America. Venom toxins can cause severe local damage and multi-organ dysfunction, impacting the neurological, hematological, and vascular systems. Endocrine disorders, though less frequently reported, can occur, with anterior pituitary insufficiency being the most common. This is typically found following bite from Russell’s viper (Daboia russelii and D. siamensis). The presentation of hypopituitarism can be acute or delayed (147).

Pathophysiology is similar to Sheehan’s syndrome and results from hemorrhagic infarction in an engorged gland, made susceptible by venom toxin. Kidney injury and disseminated intravascular coagulation (DIC) are predictors of the development of hypopituitarism. Pituitary imaging may show a spectrum of findings from completely normal to an empty sella (148). Hypogonadotropic hypogonadism may present as erectile dysfunction. Delayed puberty has been reported in males (149). The interested reader may refer to the Endotext chapter “Snakebite Envenomation and Endocrine Dysfunction” for further details (150).

CHRONIC SYSTEMIC DISEASES

The prevalence of diabetes and metabolic syndrome in tropical countries has been rising significantly in recent years (151). Type 2 diabetes in tropical countries shows distinctive features such as onset at younger ages and lower levels of obesity compared to Caucasians (152). Functional hypogonadotropic hypogonadism (FHH) has emerged as an important complication of diabetes, obesity, and metabolic syndrome across the globe. FHH results from impaired HPG axis function in the absence of an organic cause, leading to decreased testosterone levels, low or normal gonadotropin levels, and subfertility or infertility (153).

 In a study from China, 26% of men with diabetes had hypogonadotropic hypogonadism and its presence correlated with BMI (154). Lifestyle changes and weight loss can improve insulin sensitivity and restore normal HPG axis function. Testosterone replacement therapy (TRT) may be indicated in some men, although it should be used cautiously and monitored for potential side effects. Optimizing diabetes management and treating obesity are crucial and may improve hypogonadal status (155).

FHH can coexist in individuals with malnutrition and chronic energy deficit, malignancy, chronic opioid exposure, chronic kidney disease, chronic liver disease, rheumatoid arthritis, chronic obstructive pulmonary disease, depression, and other psychiatric disorders. Systemic diseases can downregulate GnRH secretion by the hypothalamus and lead to secondary hypogonadism. This is thought to be at least partly due to the direct effects of elevated inflammatory cytokines, such as IL-1, IL-6, and TNFα (156). Sickle cell disease can cause vaso-occlusive crises and can induce both primary and/or secondary hypogonadism (157,158).

 

The misuse of anabolic steroids and other hormones for performance enhancement is described among athletes and bodybuilders. Chronic abuse of these hormones can disrupt normal endocrine function, leading to hypogonadism, testicular atrophy, gynecomastia, and infertility (159).

Impairment of sperm characteristics, including alteration in total number, concentration, motility, normal morphology, prostate gland hyperplasia, and hypertrophy are recognized (160). Androgen abuse can lead to hypogonadotropic hypogonadism also, as it negatively impacts the HPG axis (161). The adverse effects may reverse over 6-18 months after discontinuation, although testicular volume and SHBG levels may not fully recover. There can be persistent quantitative and qualitative sperm changes 8–30 weeks following withdrawal of anabolic steroids (162).

The use of recreational drugs, including cannabis and opioids, has been linked to negative effects on male reproductive health. Studies have shown that these substances can decrease sperm quality, increase sperm DNA fragmentation, and lower fertility in men (163,164). Heavy use of cannabis (marijuana) has been associated with reduced semen quality, potentially due to disruption of the endocannabinoid system (ECS) in the male reproductive tract by exogenous cannabinoids. The ECS is crucial in regulating various physiological processes, including reproduction. Exogenous cannabinoids from marijuana may interfere with the normal functioning of the ECS, leading to negative effects on semen quality (165). Additionally, opioids have been found to induce secondary hypogonadism by suppressing the activity of kisspeptin-neurokinin B-dynorphin neurons. They may directly affect the testes, through endogenous opioid receptors present there (166).

CHALLENGES TO MANAGEMENT IN TROPICS

 

Male gonadal disorders in the tropics face unique challenges due to a combination of healthcare, socioeconomic, and environmental factors. These include inadequate healthcare infrastructure, especially in rural areas, economic constraints with high costs of diagnosis and treatment, and limited awareness among the population and healthcare providers, leading to underdiagnosis. Further, the cultural stigmas and beliefs around sexual health deterring men from seeking help, deficiencies in training of primary care providers to diagnose and manage gonadal disorders, complications from the tropical climate, and the high burden of infectious diseases add to the problem. There is also a scarcity of treatment guidelines tailored to regional needs and inadequate research and evidence to guide therapy.

These challenges necessitate comprehensive strategies that address healthcare infrastructure improvements, affordability, awareness campaigns, cultural sensitivity training, enhanced medical education, research into tropical-specific treatments, and telemedicine utilization for remote areas. All require collaboration among various stakeholders to improve hypogonadism management in tropical regions.

 

CONCLUSION

 

Male hypogonadism in the tropics is caused by a combination of factors, including high prevalence of infectious diseases, exposure to environmental toxins, chronic heat stress, systemic disorders including diabetes and obesity, nutritional deficiencies, and substance abuse. Significant challenges exist due to limited healthcare access, high costs, low awareness, cultural stigma, inadequate training for primary care providers, environmental factors, and a lack of region-specific treatment guidelines. These issues lead to underdiagnosis and poor management of male hypogonadism in the tropics. Improving healthcare infrastructure, raising awareness, enhancing provider training, and developing tailored treatment guidelines are essential to address these challenges effectively.

REFERENCES

  1. O’Donnell L, Stanton P, de Kretser DM. Endocrinology of the Male Reproductive System and Spermatogenesis. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 2]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279031/
  2. Thirumalai A, Anawalt BD. Epidemiology of Male Hypogonadism. Endocrinol Metab Clin North Am. 2022 Mar;51(1):1–27.
  3. Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reprod Biol Endocrinol. 2015 Apr 26;13:37.
  4. Richens J. Genital manifestations of tropical diseases. Sex Transm Infect. 2004 Feb;80(1):12–7.
  5. Liu W, Han R, Wu H, Han D. Viral threat to male fertility. Andrologia. 2018 Dec;50(11):e13140.
  6. Pellati D, Mylonakis I, Bertoloni G, Fiore C, Andrisani A, Ambrosini G, et al. Genital tract infections and infertility. Eur J Obstet Gynecol Reprod Biol. 2008 Sep;140(1):3–11.
  7. WHO Manual for the Standardized Investigation and Diagnosis of the Infertile Male [Internet]. [cited 2024 May 29]. Available from: https://www.who.int/publications-detail-redirect/9780521774741
  8. Zhao S, Zhu W, Xue S, Han D. Testicular defense systems: immune privilege and innate immunity. Cell Mol Immunol. 2014 Sep;11(5):428–37.
  9. Filippini A, Riccioli A, Padula F, Lauretti P, D’Alessio A, De Cesaris P, et al. Immunology and immunopathology of the male genital tract: Control and impairment of immune privilege in the testis and in semen. Human Reproduction Update. 2001 Sep 1;7(5):444–9.
  10. Beard CM, Benson RC, Kelalis PP, Elveback LR, Kurland LT. The incidence and outcome of mumps orchitis in Rochester, Minnesota, 1935 to 1974. Mayo Clin Proc. 1977 Jan;52(1):3–7.
  11. Adamopoulos DA, Lawrence DM, Vassilopoulos P, Contoyiannis PA, Swyer GI. Pituitary-testicular interrelationships in mumps orchitis and other viral infections. Br Med J. 1978 May 6;1(6121):1177–80.
  12. Wu H, Wang F, Tang D, Han D. Mumps Orchitis: Clinical Aspects and Mechanisms. Front Immunol. 2021 Mar 18;12:582946.
  13. De Vincentis S, Rochira V. Update on acquired hypogonadism in men living with HIV: pathogenesis, clinic, and treatment. Front Endocrinol (Lausanne). 2023 Jun 26;14:1201696.
  14. Santi D, Spaggiari G, Vena W, Pizzocaro A, Maggi M, Rochira V, et al. The Prevalence of Hypogonadism and the Effectiveness of Androgen Administration on Body Composition in HIV-Infected Men: A Meta-Analysis. Cells. 2021 Aug 12;10(8):2067.
  15. Lachâtre M, Pasquet A, Ajana F, Soudan B, Quertainmont Y, Lion G, et al. Hypogonadism: a neglected comorbidity in young and middle-aged HIV-positive men on effective combination antiretroviral therapy. AIDS. 2022 Jul 1;36(8):1061.
  16. Iddi S, Dika H, Kidenya BR, Kalluvya S. Serum gonadal hormones levels and hypogonadism in ART naïve newly diagnosed HIV infected adult males in Mwanza, Tanzania. BMC Endocr Disord. 2024 Apr 23;24:50.
  17. Wong WY, Zielhuis GA, Thomas CMG, Merkus HMWM, Steegers-Theunissen RPM. New evidence of the influence of exogenous and endogenous factors on sperm count in man. Eur J Obstet Gynecol Reprod Biol. 2003 Sep 10;110(1):49–54.
  18. Yuan J. Genitourinary Presentation of Tuberculosis. Rev Urol. 2015;17(2):102–5.
  19. Rochira V, Guaraldi G. Hypogonadism in the HIV-infected man. Endocrinol Metab Clin North Am. 2014 Sep;43(3):709–30.
  20. Collazos J, Ibarra S, Martínez E, Mayo J. Serum prolactin concentrations in patients infected with human immunodeficiency virus. HIV Clin Trials. 2002;3(2):133–8.
  21. Aggarwal J, Taneja RS, Gupta PK, Wali M, Chitkara A, Jamal A. Sex hormone Profile in Human Immunodeficiency Virus-Infected Men and It’s Correlation with CD4 Cell Counts. Indian J Endocrinol Metab. 2018;22(3):328–34.
  22. Biglia A, Blanco JL, Martínez E, Domingo P, Casamitjana R, Sambeat M, et al. Gynecomastia among HIV-infected patients is associated with hypogonadism: a case-control study. Clin Infect Dis. 2004 Nov 15;39(10):1514–9.
  23. Nuttall FQ, Warrier RS, Gannon MC. Gynecomastia and drugs: a critical evaluation of the literature. Eur J Clin Pharmacol. 2015 May;71(5):569–78.
  24. Le Tortorec A, Satie AP, Denis H, Rioux-Leclercq N, Havard L, Ruffault A, et al. Human prostate supports more efficient replication of HIV-1 R5 than X4 strains ex vivo. Retrovirology. 2008 Dec 31;5:119.
  25. De Paepe ME, Waxman M. Testicular atrophy in AIDS: a study of 57 autopsy cases. Hum Pathol. 1989 Mar;20(3):210–4.
  26. Salehian B, Jacobson D, Swerdloff RS, Grafe MR, Sinha-Hikim I, McCutchan JA. Testicular pathologic changes and the pituitary-testicular axis during human immunodeficiency virus infection. Endocr Pract. 1999;5(1):1–9.
  27. Winters SJ. Laboratory Assessment of Testicular Function. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 7]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279145/
  28. Hayes F, Dwyer A, Pitteloud N. Hypogonadotropic Hypogonadism (HH) and Gonadotropin Therapy. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 7]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279078/
  29. Shindel AW, Lue TF. Medical and Surgical Therapy of Erectile Dysfunction. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 7]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK278925/
  30. Bhasin S, Brito JP, Cunningham GR, Hayes FJ, Hodis HN, Matsumoto AM, et al. Testosterone Therapy in Men With Hypogonadism: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2018 May 1;103(5):1715–44.
  31. Musso D, Gubler DJ. Zika Virus. Clin Microbiol Rev. 2016 Jul;29(3):487–524.
  32. Joguet G, Mansuy JM, Matusali G, Hamdi S, Walschaerts M, Pavili L, et al. Effect of acute Zika virus infection on sperm and virus clearance in body fluids: a prospective observational study. Lancet Infect Dis. 2017 Nov;17(11):1200–8.
  33. Matusali G, Houzet L, Satie AP, Mahé D, Aubry F, Couderc T, et al. Zika virus infects human testicular tissue and germ cells. J Clin Invest. 2018 Oct 1;128(10):4697–710.
  34. Almeida R das N, Braz-de-Melo HA, Santos I de O, Corrêa R, Kobinger GP, Magalhaes KG. The Cellular Impact of the ZIKA Virus on Male Reproductive Tract Immunology and Physiology. Cells. 2020 Apr 18;9(4):1006.
  35. Depuydt CE, Donders GGG, Verstraete L, Vanden Broeck D, Beert JFA, Salembier G, et al. Infectious human papillomavirus virions in semen reduce clinical pregnancy rates in women undergoing intrauterine insemination. Fertil Steril. 2019 Jun;111(6):1135–44.
  36. Lyu Z, Feng X, Li N, Zhao W, Wei L, Chen Y, et al. Human papillomavirus in semen and the risk for male infertility: a systematic review and meta-analysis. BMC Infect Dis. 2017 Nov 9;17(1):714.
  37. Yas A, Mansouri Ghezelhesari E, Iranifard E, Taghipour A, Mahmoudinia M, Latifnejad Roudsari R. The Impact of Herpes Simplex Virus on Semen Parameters in Men with Idiopathic Infertility: A Systematic Review. Int J Fertil Steril. 2023;17(3):152–9.
  38. Huang J, Zhong Y, Fang X, Xie Q, Kang X, Wu R, et al. Hepatitis B virus s protein enhances sperm apoptosis and reduces sperm fertilizing capacity in vitro. PLoS One. 2013;8(7):e68688.
  39. Wang Z, Liu W, Zhang M, Wang M, Wu H, Lu M. Effect of Hepatitis B Virus Infection on Sperm Quality and Outcomes of Assisted Reproductive Techniques in Infertile Males. Front Med (Lausanne). 2021;8:744350.
  40. Brown TT. Hypogonadism in Men With Hepatitis C: What Is a Clinician to Do? Clin Infect Dis. 2019 Aug 1;69(4):577–9.
  41. Dabizzi S, Maggi M, Torcia MG. Update on known and emergent viruses affecting human male genital tract and fertility. Basic Clin Androl. 2024 Mar 14;34:6.
  42. Kalra S, Bhattacharya S, Kalhan A. Testosterone in COVID-19 – Foe, Friend or Fatal Victim? Eur Endocrinol. 2020 Oct;16(2):88–91.
  43. Nassau DE, Best JC, Kresch E, Gonzalez DC, Khodamoradi K, Ramasamy R. Impact of the SARS-CoV-2 virus on male reproductive health. BJU Int. 2022 Feb;129(2):143–50.
  44. Can Balcı MB, Can Çilesiz N. Investigation of the relationship between COVID-19 disease and semen parameters in idiopathic male infertility patients. Eur Rev Med Pharmacol Sci. 2023 Jan;27(1):378–83.
  45. Farsimadan M, Motamedifar M. Bacterial infection of the male reproductive system causing infertility. J Reprod Immunol. 2020 Nov;142:103183.
  46. Das S, Roychoudhury S, Dey A, Jha NK, Kumar D, Roychoudhury S, et al. Bacteriospermia and Male Infertility: Role of Oxidative Stress. Adv Exp Med Biol. 2022;1358:141–63.
  47. Gimenes F, Souza RP, Bento JC, Teixeira JJV, Maria-Engler SS, Bonini MG, et al. Male infertility: a public health issue caused by sexually transmitted pathogens. Nat Rev Urol. 2014 Dec;11(12):672–87.
  48. Weidner W, Ludwig M, Miller J. Therapy in male accessory gland infection--what is fact, what is fiction? Andrologia. 1998;30 Suppl 1:87–90.
  49. Trum JW, Mol BW, Pannekoek Y, Spanjaard L, Wertheim P, Bleker OP, et al. Value of detecting leukocytospermia in the diagnosis of genital tract infection in subfertile men. Fertil Steril. 1998 Aug;70(2):315–9.
  50. Boeri L, Pederzoli F, Capogrosso P, Abbate C, Alfano M, Mancini N, et al. Semen infections in men with primary infertility in the real-life setting. Fertil Steril. 2020 Jun;113(6):1174–82.
  51. Puerta Suarez J, Sanchez LR, Salazar FC, Saka HA, Molina R, Tissera A, et al. Chlamydia trachomatis neither exerts deleterious effects on spermatozoa nor impairs male fertility. Sci Rep. 2017 Apr 25;7:1126.
  52. Chemaitelly H, Majed A, Abu-Hijleh F, Blondeel K, Matsaseng TC, Kiarie J, et al. Global epidemiology of Neisseria gonorrhoeae in infertile populations: systematic review, meta-analysis and metaregression. Sex Transm Infect. 2021 Mar;97(2):157–69.
  53. Huang C, Zhu HL, Xu KR, Wang SY, Fan LQ, Zhu WB. Mycoplasma and ureaplasma infection and male infertility: a systematic review and meta-analysis. Andrology. 2015 Sep;3(5):809–16.
  54. Cheng C, Chen X, Song Y, Wang S, Pan Y, Niu S, et al. Genital mycoplasma infection: a systematic review and meta-analysis. Reprod Health. 2023 Sep 12;20(1):136.
  55. Fraczek M, Szumala-Kakol A, Jedrzejczak P, Kamieniczna M, Kurpisz M. Bacteria trigger oxygen radical release and sperm lipid peroxidation in in vitro model of semen inflammation. Fertil Steril. 2007 Oct;88(4 Suppl):1076–85.
  56. Mohta A, Agrawal A, Sharma P, Singh A, Garg S, Kushwaha RK, et al. Endocrinological Testicular Dysfunction in Patients with Lepromatous Leprosy and the Impact of Disease on Patient’s Quality of Life. Indian Dermatol Online J. 2020;11(6):959–64.
  57. Gunawan H, Achdiat PA, Rahardjo RM, Hindritiani R, Suwarsa O. Frequent testicular involvement in multibacillary leprosy. Int J Infect Dis. 2020 Jan;90:60–4.
  58. Aggrawal K, Madhu SV, Aggrawal K, Kannan AT. Hypogonadism in male Leprosy patients--a study from rural Uttar pradesh. J Commun Dis. 2005 Sep;37(3):219–25.
  59. Morley JE, Distiller LA, Sagel J, Kok SH, Kay G, Carr P, et al. Hormonal changes associated with testicular atrophy and gynaecomastia in patients with leprosy. Clin Endocrinol (Oxf). 1977 Apr;6(4):299–303.
  60. Figueiredo AA, Lucon AM. Urogenital tuberculosis: update and review of 8961 cases from the world literature. Rev Urol. 2008;10(3):207–17.
  61. Jacob JT, Nguyen TML, Ray SM. Male genital tuberculosis. Lancet Infect Dis. 2008 May;8(5):335–42.
  62. Gorse GJ, Belshe RB. Male genital tuberculosis: a review of the literature with instructive case reports. Rev Infect Dis. 1985;7(4):511–24.
  63. Muneer A, Macrae B, Krishnamoorthy S, Zumla A. Urogenital tuberculosis — epidemiology, pathogenesis and clinical features. Nat Rev Urol. 2019 Oct;16(10):573–98.
  64. Kulchavenya E. Best practice in the diagnosis and management of urogenital tuberculosis. Ther Adv Urol. 2013 Jun;5(3):143–51.
  65. Genkil JS, Ahsun S, Mohan N, Anastasopoulou C. FRI330 A Rare Case Of Hypogonadotropic Hypogonadism In A Patient With Disseminated Tberculosis And Tuberculoma Involving Tuberculum Sellae. J Endocr Soc. 2023 Oct 5;7(Suppl 1):bvad114.1265.
  66. Bini EI, D’Attilio L, Marquina-Castillo B, Mata-Espinosa D, Díaz A, Marquez-Velasco R, et al. The implication of pro-inflammatory cytokines in the impaired production of gonadal androgens by patients with pulmonary tuberculosis. Tuberculosis (Edinb). 2015 Dec;95(6):701–6.
  67. Khodadadi J, Dodangeh M, Nasiri M. Brucellar epididymo-orchitis: Symptoms, diagnosis, treatment and follow-up of 50 patients in Iran. IDCases [Internet]. 2023 [cited 2024 May 14];32. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10020095/
  68. Martínez-García F, Regadera J, Mayer R, Sanchez S, Nistal M. Protozoan infections in the male genital tract. J Urol. 1996 Aug;156(2 Pt 1):340–9.
  69. Lloyd GL, Case JR, De Frias D, Brannigan RE. Trichomonas vaginalis orchitis with associated severe oligoasthenoteratospermia and hypogonadism. J Urol. 2003 Sep;170(3):924.
  70. Setian N, Andrade RSF, Kuperman H, Manna TD, Dichtchekenian V, Damiani D. Precocious puberty: an endocrine manifestation in congenital toxoplasmosis. J Pediatr Endocrinol Metab. 2002;15(9):1487–90.
  71. Massa G, Vanderschueren-Lodeweyckx M, Van Vliet G, Craen M, de Zegher F, Eggermont E. Hypothalamo-pituitary dysfunction in congenital toxoplasmosis. Eur J Pediatr. 1989 Aug;148(8):742–4.
  72. Bruhl HH, Bahn RC, Hayles AB. Sexual precocity associated with congenital toxoplasmosis. Proc Staff Meet Mayo Clin. 1958 Dec 24;33(26):682–6.
  73. Oygür N, Yilmaz G, Ozkaynak C, Güven AG. Central diabetes insipitus in a patient with congenital toxoplasmosis. Am J Perinatol. 1998 Mar;15(3):191–2.
  74. Shiadeh MN, Niyyati M, Fallahi S, Rostami A. Human parasitic protozoan infection to infertility: a systematic review. Parasitol Res. 2016 Feb;115(2):469–77.
  75. Pace D. Leishmaniasis. J Infect. 2014 Nov;69 Suppl 1:S10-18.
  76. Castro Coto A, Hidalgo Hidalgo H, Solano Aguilar E, Coto Chacón F. [Leishmaniasis of the genital organs]. Med Cutan Ibero Lat Am. 1987;15(2):145–50.
  77. Boersma A, Noireau F, Hublart M, Boutignon F, Lemesre JL, Racadot A, et al. Gonadotropic axis and Trypanosoma brucei gambiense infection. Ann Soc Belg Med Trop. 1989 Jun;69(2):127–35.
  78. Hublart M, Lagouche L, Racadot A, Boersma A, Degand P, Noireau F, et al. [Endocrine function and African trypanosomiasis. Evaluation of 79 cases]. Bull Soc Pathol Exot Filiales. 1988;81(3 Pt 2):468–76.
  79. Reincke M, Allolio B, Petzke F, Heppner C, Mbulamberi D, Vollmer D, et al. Thyroid dysfunction in African trypanosomiasis: a possible role for inflammatory cytokines. Clin Endocrinol (Oxf). 1993 Oct;39(4):455–61.
  80. Petzke F, Heppner C, Mbulamberi D, Winkelmann W, Chrousos GP, Allolio B, et al. Hypogonadism in Rhodesian sleeping sickness: evidence for acute and chronic dysfunction of the hypothalamic-pituitary-gonadal axis. Fertil Steril. 1996 Jan;65(1):68–75.
  81. Lidani KCF, Andrade FA, Bavia L, Damasceno FS, Beltrame MH, Messias-Reason IJ, et al. Chagas Disease: From Discovery to a Worldwide Health Problem. Front Public Health. 2019 Jul 2;7:166.
  82. Carvalho TL, Ribeiro RD, Lopes RA. The male reproductive organs in experimental Chagas’ disease. I. Morphometric study of the vas deferens in the acute phase of the disease. Exp Pathol. 1991;41(4):203–14.
  83. Rocha A, Miguel OF, Barbosa HM, Candelori I, da Silva AM, Lopes ER. [The pampiniform plexus in the chronic phase of human Chagas disease: histologic assessment]. Rev Soc Bras Med Trop. 2000;33(5):413–6.
  84. Lamano Carvalho TL, Ferreira AL, Sahão MA. [Changes in the human testis in Chagas’ disease. I. Evaluation of the kinetics of the spermatogenesis]. Rev Inst Med Trop Sao Paulo. 1982;24(4):205–13.
  85. González FB, Villar SR, Pacini MF, Bottasso OA, Pérez AR. Immune-neuroendocrine and metabolic disorders in human and experimental T. cruzi infection: New clues for understanding Chagas disease pathology. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2020 Mar 1;1866(3):165642.
  86. Prasetyo RH. Scrotal abscess, a rare case of extra intestinal amoebiasis. Trop Biomed. 2015 Sep;32(3):494–6.
  87. Abdolrasouli A, de Vries HJC, Hemmati Y, Roushan A, Hart J, Waugh MA. Sexually transmitted penile amoebiasis in Iran: a case series. Sex Transm Infect. 2012 Dec;88(8):585–8.
  88. Virmani SK. Falciparum malaria presenting as testicular pain and swelling--a rejoinder. J Assoc Physicians India. 1988 Apr;36(4):295.
  89. Muehlenbein MP, Alger J, Cogswell F, James M, Krogstad D. The reproductive endocrine response to Plasmodium vivax infection in Hondurans. Am J Trop Med Hyg. 2005 Jul;73(1):178–87.
  90. Giannopoulos A, Giamarellos-Bourboulis EJ, Adamakis I, Georgopoulou I, Petrikkos G, Katsilambros N. Epididymitis Caused by Candida glabrata: A novel infection in diabetic patients? Diabetes Care. 2001 Nov 1;24(11):2003–4.
  91. Jenkin GA, Choo M, Hosking P, Johnson PDR. Candidal Epididymo-Orchitis: Case Report and Review. Clinical Infectious Diseases. 1998 Apr 1;26(4):942–5.
  92. Nagy B, Sutka P. Demonstration of antibodies against Candida guilliermondii var. guilliermondii in asymptomatic infertile men. Mycoses. 1992;35(9–10):247–50.
  93. Eickenberg H-Unull, Amin M, Lich R. Blastomycosis of the genitourinary tract. J Urol. 1975 May;113(5):650–2.
  94. Tichindelean C, East JW, Sarria JC. Disseminated histoplasmosis presenting as granulomatous epididymo-orchitis. Am J Med Sci. 2009 Sep;338(3):238–40.
  95. Staib F, Seibold M, L’age M, Heise W, Skörde J, Grosse G, et al. Cryptococcus neoformans in the seminal fluid of an AIDS patient. A contribution to the clinical course of cryptococcosis. Mycoses. 1989 Apr;32(4):171–80.
  96. Staib F Seibold M L’age M et al. Cryptococcus neoformans in the seminal fluid of an AIDS patient: a contribution to the clinical course of cryptococcosis. Mycoses. 1989; 32: 171-180.
  97. Zinedine A, Soriano JM, Moltó JC, Mañes J. Review on the toxicity, occurrence, metabolism, detoxification, regulations and intake of zearalenone: an oestrogenic mycotoxin. Food Chem Toxicol. 2007 Jan;45(1):1–18.
  98. Pekic S, Miljic D, Popovic V. Infections of the Hypothalamic-Pituitary Region. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Mar 12]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK532083/
  99. Moore LA, Erstine EM, Prayson RA. Pituitary aspergillus infection. J Clin Neurosci. 2016 Jul;29:178–80.
  100. Strickland BA, Pham M, Bakhsheshian J, Carmichael J, Weiss M, Zada G. Endoscopic Endonasal Transsphenoidal Drainage of a Spontaneous Candida glabrata Pituitary Abscess. World Neurosurg. 2018 Jan;109:467–70.
  101. Sano T, Kovacs K, Scheithauer BW, Rosenblum MK, Petito CK, Greco CM. Pituitary pathology in acquired immunodeficiency syndrome. Arch Pathol Lab Med. 1989 Sep;113(9):1066–70.
  102. Scanarini M, Rotilio A, Rigobello L, Pomes A, Parenti A, Alessio L. Primary intrasellar coccidioidomycosis simulating a pituitary adenoma. Neurosurgery. 1991 May;28(5):748–51.
  103. Stalldecker G, Molina HA, Antelo N, Arakaki T, Sica RE, Basso A. [Hypopituitarism caused by colonic carcinoma metastasis associated with hypophysial aspergillosis]. Medicina (B Aires). 1994;54(3):248–52.
  104. Ouyang T, Zhang N, Wang L, Jiao J, Zhao Y, Li Z, et al. Primary Aspergillus sellar abscess simulating pituitary tumor in immunocompetent patient. J Craniofac Surg. 2015 Mar;26(2):e86-88.
  105. Roure S, Vallès X, Pérez-Quílez O, López-Muñoz I, Chamorro A, Abad E, et al. Male genitourinary schistosomiasis-related symptoms among long-term Western African migrants in Spain: a prospective population-based screening study. Infectious Diseases of Poverty. 2024 Mar 7;13(1):23.
  106. Kayuni S, Lampiao F, Makaula P, Juziwelo L, Lacourse EJ, Reinhard-Rupp J, et al. A systematic review with epidemiological update of male genital schistosomiasis (MGS): A call for integrated case management across the health system in sub-Saharan Africa. Parasite Epidemiol Control. 2018 Nov 23;4:e00077.
  107. Kini S, Dayoub N, Raja A, Pickering S, Thong J. Schistosomiasis-induced male infertility. Case Rep. 2009;2009:bcr0120091481.
  108. Saad AH, Abdelbaky A, Osman AM, Abdallah KF, Salem D. Possible role of Schistosoma mansoni infection in male hypogonadism. J. Egypt. Soc. Parasitol. 1999;29(2):307–323.
  109. Panda DK, Mohapatra DP. Bancroftian filariasis associated with male sterility. BMJ Case Rep. 2018;2018:bcr-2017-223236.
  110. Guiton R, Drevet JR. Viruses, bacteria and parasites: infection of the male genital tract and fertility. Basic Clin Androl. 2023 Jul 20;33:19.
  111. Ekwere PD. Filarial orchitis: a cause of male infertility in the tropics--case report from Nigeria. Cent Afr J Med. 1989 Aug;35(8):456–60.
  112. Lansoud-Soukate J, Dupont A, De Reggi ML, Roelants GE, Capron A. Hypogonadism and ecdysteroid production in Loa loa and Mansonella perstans filariasis. Acta Trop. 1989 Jul;46(4):249–56.
  113. Anne B, Raphael R. Endocrine Disruptor Chemicals. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 23]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK569327/
  114. Vandenberg LN. Toxicity testing and endocrine disrupting chemicals. Adv Pharmacol. 2021;92:35–71.
  115. de Araújo-Ramos AT, Passoni MT, Romano MA, Romano RM, Martino-Andrade AJ. Controversies on Endocrine and Reproductive Effects of Glyphosate and Glyphosate-Based Herbicides: A Mini-Review. Front Endocrinol [Internet]. 2021 Mar 15 [cited 2024 May 25];12. Available from: https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2021.627210/full
  116. Doumouchtsis KK, Doumouchtsis SK, Doumouchtsis EK, Perrea DN. The effect of lead intoxication on endocrine functions. J Endocrinol Invest. 2009 Feb 1;32(2):175–83.
  117. Sun H, Xiang P, Luo J, Hong H, Lin H, Li HB, et al. Mechanisms of arsenic disruption on gonadal, adrenal and thyroid endocrine systems in humans: A review. Environment International. 2016 Oct 1;95:61–8.
  118. Sharma BM, Bharat GK, Tayal S, et al. The legal framework to manage chemical pollution in India and the lesson from the persistent organic pollutants (POPs) Sci Total Environ. 2014;490:733–747.
  119. Babu-Rajendran R, Preethi G, Poopal RK, et al. GC–MS determination of phthalate esters in human urine: a potential biomarker for phthalate bio-monitoring. J Chromatogr B. 2018;1079:15–24.
  120. Mukherjee Das A, Gogia A, Garg M, et al. Urinary concentration of endocrine-disrupting phthalates and breast cancer risk in Indian women: a case-control study with a focus on mutations in phthalate-responsive genes. Cancer Epidemiol. 2022;79:102188. doi: 10.1016/j.canep.2022.102188.
  121. Eskenazi B, An S, Rauch SA, Coker ES, Maphula A, Obida M, et al. Prenatal Exposure to DDT and Pyrethroids for Malaria Control and Child Neurodevelopment: The VHEMBE Cohort, South Africa. Environmental Health Perspectives [Internet]. 2018 Apr [cited 2024 May 25];126(4). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6071803/
  122. Jeng HA. Exposure to Endocrine Disrupting Chemicals and Male Reproductive Health. Front Public Health. 2014 Jun 5;2:55.
  123. Sharma A, Mollier J, Brocklesby RWK, Caves C, Jayasena CN, Minhas S. Endocrine-disrupting chemicals and male reproductive health. Reprod Med Biol. 2020 Jul;19(3):243–53.
  124. Lahimer M, Abou Diwan M, Montjean D, Cabry R, Bach V, Ajina M, et al. Endocrine disrupting chemicals and male fertility: from physiological to molecular effects. Front Public Health. 2023 Oct 10;11:1232646.
  125. Jaeger C, Allend€orfer J, Hatziagelaki E, Dyrberg T, Bergis K, Federlin K, et al. Persistent GAD 65 Antibodies in Longstanding IDDM are not Associated with Residual Beta-Cell Function, Neuropathy or HLA-DR Status. Horm Metab Res 1997; 29: 510–515.
  126. Li DK, Zhou Z, Miao M, et al. Urine bisphenol A (BPA) level in relation to semen quality. Fertil Steril 2011;95:625–30.
  127. Skakkebaek NE. Testicular dysgenesis syndrome. Horm Res. 2003;60 Suppl 3:49.
  128. Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects. Hum Reprod. 2001 May;16(5):972–8.
  129. Cargnelutti F, Di Nisio A, Pallotti F, Sabovic I, Spaziani M, Tarsitano MG, et al. Effects of endocrine disruptors on fetal testis development, male puberty, and transition age. Endocrine. 2021;72(2):358–74.
  130. Durmaz E, Ozmert EN, Erkekoglu P, Giray B, Derman O, Hincal F, et al. Plasma phthalate levels in pubertal gynecomastia. Pediatrics. 2010 Jan;125(1):e122-129.
  131. Brody SA, Loriaux DL. Epidemic of gynecomastia among haitian refugees: exposure to an environmental antiandrogen. Endocr Pract. 2003;9(5):370–5.
  132. Ramsey JT, Li Y, Arao Y, Naidu A, Coons LA, Diaz A, et al. Lavender Products Associated With Premature Thelarche and Prepubertal Gynecomastia: Case Reports and Endocrine-Disrupting Chemical Activities. J Clin Endocrinol Metab. 2019 Nov 1;104(11):5393–405.
  133. Hansen J. Elevated risk for male breast cancer after occupational exposure to gasoline and vehicular combustion products. Am J Ind Med. 2000 Apr;37(4):349–52.
  134. Szabo GK, Vandenberg LN. REPRODUCTIVE TOXICOLOGY: The male mammary gland: a novel target of endocrine-disrupting chemicals. Reproduction. 2021 Nov 1;162(5):F79–89.
  135. Koifman S, Koifman RJ, Meyer A. Human reproductive system disturbances and pesticide exposure in Brazil. Cad Saude Publica. 2002;18(2):435–45.
  136. McGlynn KA, Quraishi SM, Graubard BI, Weber JP, Rubertone MV, Erickson RL. Persistent organochlorine pesticides and risk of testicular germ cell tumors. J Natl Cancer Inst. 2008 May 7;100(9):663–71.
  137. Biggs ML, Davis MD, Eaton DL, Weiss NS, Barr DB, Doody DR, et al. Serum organochlorine pesticide residues and risk of testicular germ cell carcinoma: a population-based case-control study. Cancer Epidemiol Biomarkers Prev. 2008 Aug;17(8):2012–8.
  138. Bhattacharya S, Sahay R, Afsana F, Sheikh A, Widanage NM, Maskey R, et al. Global Warming and Endocrinology: The Hyderabad Declaration of the South Asian Federation of Endocrine Societies. Indian Journal of Endocrinology and Metabolism. 2024 Apr;28(2):129.
  139. Hoang-Thi AP, Dang-Thi AT, Phan-Van S, Nguyen-Ba T, Truong-Thi PL, Le-Minh T, et al. The Impact of High Ambient Temperature on Human Sperm Parameters: A Meta-Analysis. Iran J Public Health. 2022 Apr;51(4):710–23.
  140. Barratt CLR, Björndahl L, De Jonge CJ, et al. The diagnosis of male infertility: an analysisof the evidence to support the development of global WHO guidance-challenges andfuture research opportunities. Hum Reprod Update 2017; 23:660.
  141. Bonde JP, Giwercman A, Ernst E. Identifying environmental risk to male reproductive function by occupational sperm studies: logistics and design options. Occup Environ Med. 1996 Aug;53(8):511–9.
  142. Wang C, Cui YG, Wang XH, et al. transient scrotal hyperthermia and levonorgestrelenhance testosterone-induced spermatogenesis suppression in men through increasedgerm cell apoptosis. J Clin Endocrinol Metab 2007.
  143. Thonneau P, Ducot B, Bujan L, et al. Effect of male occupational heat exposure on timeto pregnancy. Int J Androl 1997; 20:274.
  144. Bolt JW, Evans C, Marshall VR. Sexual dysfunction after prostatectomy. Br J Urol 1987;59:319.
  145. Copuroglu C, Yilmaz B, Yilmaz S, et al. Sexual dysfunction of male, after pelvic fracture.Eur J Trauma Emerg Surg 2017; 43:59.
  146. Hari Kumar KV, Swamy MN, Khan MA. Prevalence of hypothalamo pituitary dysfunction in patients of traumatic brain injury. Indian J Endocrinol Metab. 2016 Nov-Dec;20(6):772-778.
  147. Bhattacharya S, Krishnamurthy A, Gopalakrishnan M, Kalra S, Kantroo V, Aggarwal S, et al. Endocrine and Metabolic Manifestations of Snakebite Envenoming. Am J Trop Med Hyg. 2020 Oct;103(4):1388–96.
  148. Yerawar C, Punde D, Pandit A, Deokar P. Russell’s viper bite and the empty sella syndrome. QJM Mon J Assoc Physicians. 2021 Jul 28;114(4):255–7.
  149. Shivaprasad C, Aiswarya Y, Sridevi A, Anupam B, Amit G, Rakesh B, et al. Delayed hypopituitarism following Russell’s viper envenomation: a case series and literature review. Pituitary. 2019 Feb;22(1):4–12.
  150. Bhattacharya S, Nagendra L, Tyagi P. Snakebite Envenomation and Endocrine Dysfunction. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 26]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK575924/
  151. Unnikrishnan R, Mohan V. Diabetes in the tropics: prevalent, increasing and a major public health problem. Trans R Soc Trop Med Hyg. 2016 May;110(5):263–4.
  152. Kapoor N. Thin Fat Obesity: The Tropical Phenotype of Obesity. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2024 May 27]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK568563/
  153. Spaziani M, Carlomagno F, Tarantino C, Angelini F, Vincenzi L, Gianfrilli D. New perspectives in functional hypogonadotropic hypogonadism: beyond late onset hypogonadism. Front Endocrinol (Lausanne). 2023;14:1184530.
  154. Zhou Y, Tian R, Wang X, Sun J, Zhu L, An X. The occurrence of hypogonadotropic hypogonadism in Chinese men with type 2 diabetes. Clinical Endocrinology. 2022;96(6):837–46.
  155. Corona G, Rastrelli G, Morelli A, Sarchielli E, Cipriani S, Vignozzi L, et al. Treatment of Functional Hypogonadism Besides Pharmacological Substitution. World J Mens Health. 2020 Jul;38(3):256–70.
  156. Esquivel-Zuniga R, Rogol AD. Functional hypogonadism in adolescence: an overlooked cause of secondary hypogonadism. Endocrine Connections [Internet]. 2023 Nov 1 [cited 2024 May 28];12(11). Available from: https://ec.bioscientifica.com/view/journals/ec/12/11/EC-23-0190.xml
  157. Musicki B, Burnett AL. Testosterone Deficiency in Sickle Cell Disease: Recognition and Remediation. Front Endocrinol (Lausanne). 2022 May 3;13:892184.
  158. Ribeiro APMR, Silva CS, Zambrano JCC, Miranda J de OF, Molina CAF, Gomes CM, et al. Compensated hypogonadism in men with sickle cell disease. Clinical Endocrinology. 2021;94(6):968–72.
  159. Solanki P, Eu B, Smith J, Allan C, Lee K. Physical, psychological and biochemical recovery from anabolic steroid-induced hypogonadism: a scoping review. Endocrine Connections [Internet]. 2023 Dec 1 [cited 2024 May 28];12(12). Available from: https://ec.bioscientifica.com/view/journals/ec/12/12/EC-23-0358.xml
  160. El Osta R, Almont T, Diligent C, Hubert N, Eschwège P, Hubert J. Anabolic steroids abuse and male infertility. Basic Clin Androl. 2016 Feb 6;26:2.
  161. Dohle GR, Smit M, Weber RF. Androgens and male fertility. World J Urol. 2003;21(5):341–345.
  162. McBride JA, Coward RM. Recovery of spermatogenesis following testosterone replacement therapy or anabolic-androgenic steroid use. Asian J Androl. 2016;18(3):373–80.
  163. Bracken MB, Eskenazi B, Sachse K, McSharry JE, Hellenbrand K, Leo-Summers L. Association of cocaine use with sperm concentration, motility, and morphology. Fertil Steril. 1990;53(2):315–322.
  164. Safarinejad MR, Asgari SA, Farshi A, et al. The effects of opiate consumption on serum reproductive hormone levels, sperm parameters, seminal plasma antioxidant capacity and sperm DNA integrity. Reprod Toxicol. 2013;36:18–23.
  165. Nielsen JE, Rolland AD, Rajpert-De Meyts E, Janfelt C, Jørgensen A, Winge SB, et al. Characterisation and localisation of the endocannabinoid system components in the adult human testis. Sci Rep. 2019 Sep 19;9(1):12866.
  166. Subirán N, Casis L, Irazusta J. Regulation of male fertility by the opioid system. Mol Med. 2011;17(7–8): 846–853.

Adrenal Incidentalomas

ABSTRACT

 

Wider application and technical improvement of abdominal imaging procedures in recent years, has led to the discovery of unsuspected adrenal tumors in an increasing frequency. These incidentally detected lesions, also called adrenal incidentalomas, have become a common clinical problem and need to be investigated for evidence of hormonal hypersecretion and/or malignancy. In this chapter, current information on the prevalence, etiology, radiological features, and appropriate biochemical evaluation are presented as a narrative review of the available literature. Despite the flurry of data accumulated, controversies are still present regarding the accuracy of diagnostic tests and cut-offs utilized to establish hormonal hypersecretion, potential long-term sequelae, indications for surgical treatment as well as duration and intensity of conservative management and follow-up. Recently, clinical guidelines proposing diagnostic and therapeutic algorithms have been published to aid in clinical practice, however an individualized approach through a multidisciplinary team of experts is recommended.

 

INTRODUCTION

 

Abdominal computed tomography (CT), since its introduction in the late 1970’s, has proven to be an excellent tool for identifying pathology in patients with suspected adrenal disease. It was also predicted that the ability of CT to image both adrenal glands could lead to the occasional discovery of asymptomatic adrenal disease (1). Nowadays, further technological advances and broader availability of CT and other imaging modalities such as Ultrasonography (US), Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) have made the detection of unexpected lesions in adrenal and other endocrine glands a common finding (2). Although incidental detection of adrenal disease may lead to earlier diagnosis and possibly improved outcome in certain cases, it is now recognized that diagnostic evaluation and follow-up of all incidentally discovered adrenal masses, or so-called “adrenal incidentalomas”, may put a significant burden on patient’s anxiety and health and produce increasing financial consequences for the health system (3). It is therefore important to develop cost-effective strategies to diagnose and manage patients with adrenal incidentalomas.

 

DEFINITION

 

According to the NIH State-of-the-Science Statement (4), adrenal incidentalomas (AIs) are defined as clinically inapparent adrenal masses discovered serendipitously during diagnostic testing or treatment for conditions not related to the adrenals, such as abdominal or back pain or for exclusion of pulmonary embolism or other lung disease. Although an arbitrary cut-off of 1 cm or more has been employed to define an adrenal lesion as AI (5,6), this cut-off might be challenged following the higher resolution that modern imaging modalities offer, mainly MRI and CT. Nonetheless, in all published guidelines this cut-off is accepted as the minimum size above which additional diagnostic work-up should be performed, unless clinical signs and symptoms suggestive of adrenal hormone excess are present. Patients harboring an AI, by definition, should not have any history, signs, or symptoms of adrenal disease prior to the imaging procedure that led to its discovery. This strict definition excludes cases in which adrenal-specific signs and symptoms are “missed” during history taking or physical examination, or in which a hereditary syndrome associated with an increased likelihood to develop adrenal tumors is suspected (6). Similarly, adrenal masses discovered on imaging for tumor staging or follow-up in extra-adrenal malignancies fall outside the definition of an AI (7). This is because adrenal metastases are a common finding in this setting, with a prevalence ranging from 3 to 40% in autopsy and from 6 to 20% in radiological series (8). A recent population-based cohort study reported a 22-fold higher likelihood of an AI being a metastatic lesion when discovered during cancer staging, reaching a prevalence of 7.5% (9). In another single-center cohort study including 475 patients with colorectal cancer, the incidence of AIs was 10.5% (10).

 

EPIDEMIOLOGY

 

The precise prevalence and incidence of AIs cannot be easily defined since data from population-based studies are scarce. Most previous data were extrapolated from autopsy or radiological studies that are inherently biased due to their retrospective nature, insufficient clinical information, different referral patterns and patient selection criteria.

 

In autopsy studies, the reported prevalence of AIs was found to be around 2.3%, ranging from 1 to 8.7% (11–23), without any significant gender difference. The prevalence of AIs increases with age, being 0.2% in young subjects compared to 6.9% in subjects older than 70 years of age (24), and is higher in white, obese, diabetic, and hypertensive patients (8). The variability of the reported prevalence in different series could also be attributed to the size cut-off used for the definition of AI as in some post-mortem series, small nodules (<1 cm) were detected in more than half of the patients examined (23).

 

In radiological studies, the prevalence of AIs differs depending on the imaging modality used and should be interpreted carefully due to referral and under-reporting bias. Transabdominal US during a routine health examination identified AIs in 0.1% of those screened (25), while studies using CT reported a mean prevalence of 0.64% ranging from 0.35 to 1.9% in a total of 82,483 scans published in the literature between 1982 and 1994 (21,26–30). However, two more recent studies utilizing high-resolution CT scanning technology, have reported prevalence rates of 4.4% and 5% respectively, which are more consistent with those observed in autopsy studies (31,32). This increase in detection frequency paralleled by the technological advances in medical imaging quality, can explain why AIs are considered a “disease of modern technology”. Age has also been found to affect AI radiological detection rates, as these lesions are found in 0.2% of individuals younger than 30 years, in 3% at the age of 50 years and up to 10% in individuals above 70 years of age (24,31,33). However, a recent publication from China including 25,356 healthy individuals (aged 18-78) who underwent abdominal CT imaging as part of a funded health check, reported an AI detection rate of 1.4%, increasing with age, from 0.2% in the youngest group (18-25 years) to 3.2% in those older than 65 years (34). The prevalence of AIs is very low in childhood and adolescence accounting for 0.3-0.4% of all tumors (35). Adrenal incidentalomas appear to be slightly more frequent in women in radiological series, in discordance with autopsy studies, probably because women undergo abdominal imaging more frequently than men (33). Bilateral AIs are found in 10-15% of cases (36), while distribution between the two adrenals appears to be similar in both post-mortem and CT studies (8,33).

 

DIFFERENTIAL DIAGNOSIS

 

Adrenal Incidentalomas are not a single pathological entity, but rather comprise a spectrum of different pathologies that share the same path of discovery and include both benign and malignant lesions arising from the adrenal cortex, the medulla, or being of extra-adrenal origin (Table 1).

 

Table 1. The Spectrum of Lesions Presenting as AIs, Modified from (37)

Adrenal Cortex lesions

·    Adenoma (non-functioning)

·    Adenoma (functioning)

-        Cortisol-secreting (MACS)

-        Aldosterone-secreting

·    Nodular hyperplasia (primary bilateral macronodular adrenal hyperplasia)*

·    Adrenocortical Carcinoma (secreting or non-secreting)

Adrenal Medulla lesions

·    Pheochromocytoma (benign or malignant)*

·    Ganglioneuroma

·   Neuroblastoma, ganglioneuroblastoma

Other adrenal lesions

·    Myelolipoma, lipoma

·    Hemangioma, angiosarcoma

·    Cyst

·    Hamartoma, teratoma

Metastases* (lung, breast, kidney, melanoma, lymphoma)

Infiltration*

·    Amyloidosis

·    Sarcoidosis

·    Lymphoma

Infections*

·    Abscess

·     

·    Fungal/parasitic (histoplasmosis, coccidiomycosis, tuberculosis)

·    Cytomegalovirus

Adrenal hemorrhage or hematomas*

Adrenal pseudotumors

Congenital Adrenal Hyperplasia (CAH)*

* Should be considered when bilateral adrenal lesions are detected

 

In general, the vast majority (80-90%) of AIs are benign adrenal adenomas, as shown by accumulated follow-up data from their natural history, even in the absence of pathological confirmation, since adrenal adenomas are rarely excised (5). However, a number of these lesions may be malignant and/or exhibit autonomous hormonal secretion that is not clinically detected due to subtle secretory pattern or periodical secretion. Therefore, the task a physician faces when dealing with an AI is mainly to exclude malignant and functioning tumors.

 

Mild autonomous cortisol secretion (MACS) is the most frequent endocrine dysfunction detected in patients with AIs, with a prevalence ranging from 5 to 30%, depending on the study design, work-up protocols, and mainly diagnostic criteria used (5). This condition exclusively identified in the setting of AIs, also termed subclinical Cushing’s syndrome or subclinical hypercortisolism, is characterized by the absence of the typical clinical phenotype of hypercortisolism and by the presence of subtle alterations of the hypothalamic-pituitary-adrenal (HPA) axis. These tumors do not secrete cortisol under the physiological control of corticotropin (ACTH), but rather autonomously and in some cases under the control of one or more aberrant hormone receptors (38,39).

 

Pheochromocytomas (PCCs), albeit rare in the general population, are discovered in approximately 5% of patients with AIs (40), while more than 30% of PCCs are diagnosed as AIs (41). Clinical manifestations are highly variable, and the classic clinical triad (headache, palpitations and diaphoresis) is not present in most patients. In addition, several patients harbor ‘‘silent pheochromocytomas’’, being totally asymptomatic or having intermittent and subtle symptoms. In a large multicentric study, approximately half of the patients with PCCs presenting as AIs were normotensive, whereas the remaining had mild to moderate hypertension (33).

 

Primary aldosteronism (PA) has a median prevalence of 2% (range 1.1-10%) among patients with AIs (42). After excluding cases with severe hypertension and hypokalemia a retrospective study found that 16 out of 1004 subjects with AIs (1.5%) had PA (33). This figure is relatively low when compared to the prevalence of PA in unselected hypertensive populations which ranges from 4.6 to 16.6% (43) and may be related to the different investigational protocols and cut-offs indicative of autonomous aldosterone secretion used. The absence of hypokalemia does not exclude this condition, but absence of hypertension makes PA unlikely, although normotensive patients with PA have occasionally been reported (44). A recent study using a new diagnostic approach, considering the stimulatory effect that adrenocorticotropin (ACTH) could exert on aldosterone secretion, revealed a 12% prevalence of PA in normotensive and normokalemic patients with AIs (45).

 

Over secretion of adrenal androgens is usually accompanied with clinical signs or symptoms of virilization in women and feminization in men (46), thus falling out of the strict definition of AI’s requiring absence of adrenal-related manifestations. Presence of elevated adrenal androgens should alert the physicians for the possibility of an adrenocortical carcinoma, although benign androgen-secreting tumors have rarely been reported (47).

 

Combining studies that used a broad definition of incidentaloma without clearly stated inclusion criteria and those that reported descriptions of individual cases, Mansmann et al found 41% of AIs to be adenomas, 19% metastases, 10% ACCs, 9% myelolipomas, and 8% PCCs, with other benign lesions, such as adrenal cysts, ganglioneuromas, hematomas, and infectious or infiltrative lesions representing rare pathologies (48). However, the relative prevalence of any pathology depends on the inclusion criteria used and is highly influenced by referral bias. Surgical series and data from referral centers tend to overestimate the prevalence of large, malignant and functioning tumors, because such cases are mainly referred for surgery or expert evaluation. Similarly, metastatic lesions are much more common when patients with known extra-adrenal cancer are included in the study population. The probability of an incidentally discovered adrenal lesion in a patient without a history of cancer to be metastatic is as low as 0.4% (29). Studies applying more strict inclusion criteria may identify a greater number of small and biochemically silent tumors. In a comprehensive review, Cawood et al. (3) concluded that the prevalence of malignant and functioning lesions among AIs is likely lower when strict inclusion and exclusion criteria for the study populations are used. By analyzing 9 studies that more accurately simulated the clinical scenario of a patient referred for assessment of an AI, they reported a mean prevalence of 88.1% (range 86.4-93%) for non-functioning benign adrenal adenomas (NFAIs), 6% (range 4-8.3%) for MACS, 1.2% for aldosterinomas, 1.4% (range 0.8-3%) for ACCs, 0.2% (range 0-1.4%) for metastases and 3% (range 1.8-4.3%) for PCCs. These low rates for clinically significant tumors compared to those reported by previous studies (6,8,48), highlight the limitations of epidemiological data and raise significant questions concerning the appropriate diagnostic and follow-up protocols. Notably, it has recently been suggested that a significant number of patients with small AIs do not undergo the recommended evaluation (9), adding further confusion in defining the relative prevalence of each pathology, through under-reporting bias.

 

In the case of bilateral AIs, a broader spectrum of diagnoses needs to be considered (Table 1), particularly in a relevant clinical setting, including metastatic or infiltrative diseases of the adrenals, hemorrhage, congenital adrenal hyperplasia (CAH), bilateral cortical adenomas or PCCs, and primary bilateral macronodular adrenal hyperplasia (PBMAH) (49). Occasionally, adrenal tumors of different nature may simultaneously be present in the same patient or in the same adrenal gland (50–53). Adrenal pseudotumor is a term used to describe radiological images of masses that seem to be of adrenal origin, but arise from adjacent structures, such as the kidney, spleen, pancreas, vessels and lymph nodes or are results of technical artifacts.

 

PATHOGENESIS

 

The pathogenesis of AIs is largely unknown. Early observations in autopsy studies which revealed that AIs are more frequent in older patients, led to the notion that these tumors are a manifestation of the ageing adrenal and could represent focal hyperplasia in response to ischemic injury, a concept that was supported by histopathological findings of capsular arteriopathy (54). Clonal analysis of adrenal tumors later revealed that the vast majority are of monoclonal origin and only a few arise from polyclonal focal nodular hyperplasia under the putative effect of local or extra-adrenal growth factors (55,56). In this sense, it has been postulated that hyperinsulinemia associated with the insulin resistance in individuals with the metabolic syndrome, which frequently coexists in patients harboring AIs, could contribute to the development of these tumors, through the mitogenic action of insulin on the adrenal cortex (57,58). However, the opposite causal relationship, that subtle autonomous cortisol production from AIs results in insulin resistance, has also been proposed (59). It is plausible that both pathways can be true in a reciprocal triad. Another interesting hypothesis involves alterations in the glucocorticoid feedback sensitivity of the HPA axis acting as a drive for adrenal cell proliferation especially in cases with bilateral involvement. In a recent study, unexpected ACTH and cortisol responses to the combined dexamethasone-CRH (corticotropin-releasing hormone) test were found, in about half of the patients with bilateral AIs, when compared to control and unilateral adenoma cases (60). Such a dysregulated ACTH secretion during lifetime may lead to subtle but chronic trophic stimulation of the adrenals by repeatedly inappropriately higher ACTH levels, particularly in response to stress, favoring nodular adrenal hyperplasia.

 

Although several genetic syndromes are known to be associated with adrenal tumors, germline or somatic genetic alterations are identified only in subgroups of sporadic tumors that are mainly functioning (61–63). Elucidation of specific signaling pathways involved in these familial syndromes has led to the identification of several mutations in genes not previously described in ACCs, cortisol- and aldosterone-secreting adenomas as well as PCCs, creating new insights in adrenal tumorigenesis (Figure 1). However, the genetics of benign NFAIs that account for the majority of AIs are poorly understood.

 

Figure 1. Genes Involved in the Development of Adrenocortical Tumors IN Sporadic or Familial Cases. MEN: Multiple Endocrine Neoplasia; CTNNB1: Catenin Beta-1 gene; CYP21A2: 21-Hydroxylase gene; CAH: Congenital Adrenal Hyperplasia; APC: Adenomatous polyposis coli; FAP: Familial adenomatous polyposis; KCNJ5: gene encoding potassium channel, inwardly rectifying subfamily J, member 5; ATP1A1: gene encoding sodium/potassium-transporting ATPase subunit alpha 1; ATP2B3: plasma membrane calcium-transporting ATPase 3; CACNA1D: gene encoding calcium channel, voltage-dependent, L type, alpha 1D subunit; ARMCS: Armadillo repeat containing 5; ZNRF3: gene encoding Zinc and Ring Finger3; IGF-2: Insulin-like growth factor 2; TP53: tumor protein p53; CDKN2A: cyclin-dependent kinase inhibitor 2A; RB1: retinoblastoma protein; DAXX: death-associated protein 6; GNAS: gene encoding G-protein alpha subunit: PDE11A: phosphodiesterase 11A; PDE8B: phosphodiesterase 8B; PRKACA: gene encoding catalytic subunit alpha of protein kinase A; SDH-A-B-C-D: gene encoding succinate dehydrogenase complex subunit A, B, C, and D; SDHAF2: succinate dehydrogenase complex assembly factor 2; VHL: von-Hippel-Landau; RET: rearranged during transfection proto-oncogene; MAX: myc-associated factor X; TMEM127: gene encoding transmembrane protein 127.

 

DIAGNOSTIC APPROACH

 

Although the prevalence of potentially life-threatening disorders associated with AIs is relatively low, the question of whether a lesion is malignant (mainly an ACC) or functioning needs to be addressed in patients with an incidentally discovered adrenal mass. A careful clinical examination and a detailed medical history, evaluation of the imaging characteristics of the adrenal tumor(s), and biochemical evaluation to exclude hormonal excess can help clinicians identify the few cases that pose a significant risk and intervene accordingly.

 

CLINICAL EVALUATION

 

Per definition, patients with AIs should have no signs or symptoms implying adrenal dysfunction before the radiological detection of the adrenal tumor(s). In everyday clinical practice though, physicians who are not familiar with endocrine diseases may overlook mild signs of hormone excess and pursue evaluation of adrenal function following the incidental discovery of an adrenal mass. In this setting, such cases should not be designated as AIs and highlight the need for detailed and careful clinical history and examination (64).

 

IMAGING EVALUATION

 

Distinguishing malignant from benign AI lesions should be the priority at the time of their initial detection, and determination of their imaging phenotype is currently considered the most reliable and non-invasive approach to aid in this distinction. Traditionally the size of the lesion reported by CT or MRI has been considered as indicative of malignancy as most ACCs are large or significantly larger than adenomas at the time of diagnosis (33). In a meta-analysis, ACCs represented 2% of all tumors ≤4 cm in diameter, but the risk of malignancy increased significantly with tumor size greater than 4 cm, being 6% in tumors with size 4.1-6 cm and 25% in tumors >6 cm (65). However, size alone has low specificity in distinguishing benign from malignant lesions, since ACCs can also be relatively small during early stages of development and exhibit subsequent progressive growth (5). An analysis of 4 recent studies investigating the 4cm size cut-off to distinguish benign from malignant lesions reported sensitivities ranging from 23% to 90% while the pooled sensitivity was 77% (95% CI 45%-93%) and the pooled specificity was 90% (95% CI 78%-96%) (66). Other than size, findings suggestive of malignancy include irregular shape and borders, tumor heterogeneity with central necrosis or hemorrhage, and invasion into surrounding structures. Benign adenomas are usually small (<4 cm), homogenous, with well-defined margins. Slow growth rate or stable size of an adrenal mass have also been proposed as indicators of benign nature (4). However, studies on the natural history of AIs suggest that up to 25% of benign adenomas can display increase in size by almost 1 cm, while adrenal metastases with no change in CT appearance over a period of 36 months have been described, not allowing for the introduction of a safe cut-off of absolute growth or growth rate to distinguish benign from malignant lesions (67).

 

Computed Tomography (CT)

 

CT has a high spatial and contrast resolution, which allows assessment of tissue density by measuring X-ray absorption compared to water (attenuation, expressed in Hounsfield Units - HU).  Water and air are conventionally allocated an attenuation value of 0 HU and -1000 HU respectively, while fat is usually characterized by a HU value between -40 and -100. Because there is an inverse linear relation between the fat content of a lesion and attenuation, lipid-rich adenomas express lower HU in unenhanced (without contrast medium) CT images compared to malignant lesions, which are usually lipid-poor (68). A value of ≤10 HU in unenhanced CT images is the most widely used and accepted attenuation threshold for the diagnosis of a lipid-rich, benign adrenal adenoma (69,70). In several studies a density of ≤10 HU was found to be superior to size in differentiating benign from malignant masses, displaying a sensitivity of 96-100% and a specificity of 50-100% (71). Data from 6 studies (9,72–76) on the diagnostic accuracy of unenhanced attenuation values, reported that a CT density >10 HU has a very high sensitivity for detection of adrenal malignancy (100% in all 6 studies), while the pooled specificity was clearly lower (56%-59%). This means that adrenal masses with a density of ≤10 HU are virtually never malignant, however a large number of benign lesions had HU > 10. Increasing the cutoff to HU > 20, provided a pooled sensitivity of 94%-98% and a higher specificity (75%-78%), leaving a fairly significant number of malignant tumors lying between 10 and 20 HU. In this context, the risk of malignancy in a homogeneous 5 cm adrenal mass with a CT attenuation value of 10 HU is close to 0% (49). On the other hand, up to 30-40% of benign adenomas are considered lipid-poor and have an attenuation value of >10 HU on non-contrast CT, which is considered indeterminate since it overlaps with those found in malignant lesions and PCCs. Hence, unenhanced CT attenuation is a useful screening tool to identify a lesion as benign and exclude malignancy but is less reliable in diagnosing a malignant mass with certainty. When considering patients with a history of extra-adrenal malignancy though, several studies evaluating the >10 HU cut-off as indicative of malignancy showed high sensitivity (93%) for the detection of malignancy but variable specificity, meaning that 7% of adrenal metastases were found to have a tumor density of ≤10 HU (70). Attenuation values in non-contrast CT can also reliably identify typical myelolipomas that have a density lower than -40 HU (49).

 

For those indeterminate adrenal lesions (>10HU) intravenous contrast administration reveals their hemodynamic and perfusion properties that can be utilized to distinguish benign from malignant lesions. The attenuation on delayed images (10-15 min post contrast administration) decreases more quickly in adenomas because they exhibit rapid uptake and clearance compared to malignant lesions that usually enhance rapidly but demonstrate a slower washout of contrast medium (77). There are two methods of estimating contrast medium washout: absolute percentage washout (APW) and relative percentage washout (RPW) and can be calculated from values of pre-contrast (PA), enhanced (EA, 60-70 seconds after contrast medium administration) and delayed (DA, 10-15 mins after contrast medium administration) attenuation values according to the formulas below:

 

APW=100 x (EA-DA) / (EA-PA)

RPW=100 x (EA-DA) / EA

 

Initial studies suggested that lipid-poor adenomas demonstrate rapid washout with APW >60% (sensitivity of 86-100%, specificity 83-92%) and a RPW >40% (sensitivity of 82-97%, specificity 92-100%) (78). Metastases usually demonstrate slower washout on delayed images (APW<60%, RPW<40%) than adenomas and ACCs typically have a RPW of <40% (79). It is important to note that the above values of sensitivity and specificity were produced in studies with limitations and high risk of bias due to the lack of definitive pathological diagnosis, different timing in acquiring post-contrast images, and the use of broad inclusion criteria, including not only AIs but also clinically overt adrenal masses. Recent data have suggested that these percentage washout cutoffs have suboptimal performance for characterizing benign lesions, since 22% (using APW) and 8% (using RPW) of malignant tumors are not correctly identified (70,75,80). To detect all malignant tumors, the RPW cutoff should be increased to 58%, leading to a specificity of only 15% (75).

 

Furthermore, contrast-enhanced washout CT studies may not suffice for characterization of lesions such as PCCs, cysts, and myelolipomas; in these cases, further biochemical, anatomical and/or functional imaging may be required. Findings consistent, but not diagnostic, of PCC on CT include high attenuation values, prominent vascularity, and delayed washout of contrast medium (79). Another recent study (81), showed that only a minority (21%) of cortisol-secreting adenomas has the typical unenhanced attenuation value of <10 HU, because cortisol secretion is associated with decreased intra-cytoplasmic lipid droplets containing cholesterol esters which are necessary for cortisol synthesis. Nevertheless, among the adenomas with high pre-contrast density (>10 HU), washout analysis after contrast administration was consistent with the benign nature of the tumor in 60% of the cases.

 

Another crucial key point in clinical practice is that most abdominal and chest CT scans leading to the unexpected discovery of an adrenal mass are obtained with the use of intravenous contrast that may not fulfill current technical recommendations for an optimal CT study of the adrenal glands, such as analysis on contiguous 3-5 mm-thick CT slices, preferentially on multiple sections using multidetector (MDCT) row protocols (82). In such cases, it may be worthwhile to obtain a new CT scan, specifically aimed for the study of the adrenal glands, including washout protocols in order to avoid the radiation exposure of a subsequent third CT scan in case of indeterminate unenhanced attenuation values.

 

Finally, the importance of thorough and standardized reporting by radiologists (including common terminology, nodule size, and HU) needs to be highlighted, in order to improve the percentage of patients with AIs that receive appropriate diagnostic testing and follow-up. This is a recently raised issue based on evidence that suggests that most of AIs are not adequately investigated according to international guidelines due to inconsistent use of terms and lack of specific details and recommendations in radiology reports (83–85).

 

Typical CT images of adrenal pathologies is shown in Figure 2.

 

Figure 2. CT images of adrenal pathologies presenting as adrenal incidentalomas. a,b,c: A patient with a benign (lipid-rich) adrenal adenoma with unenhanced attenuation value - 3 HU (a), early attenuation (60 seconds after i.v. contrast medium administration) 35 HU (b) and delayed attenuation (10 min post-contrast administration) 18 HU. ARW = 45% and RPW=49%. Absolute washout (APW) less than 60% is indeterminate. However, the low pre-contrast attenuation is suggestive of an adenoma. Relative washout (RPW) of 40% or higher is consistent with an adenoma; d,e,f: Biochemically and histologically proven pheochromocytoma with unenhanced attenuation of 49 HU (d), early attenuation 90 HU (e) and delayed attenuation 64 HU. ARW = 63% and RPW=29%. Absolute washout >60% is suggestive of an adenoma, however relative washout less than 40% and unenhanced attenuation >10 HU are indeterminate; g,h: A patient with a primary adrenocortical carcinoma characterized by heterogeneity an unenhanced attenuation value >10 HU (g) and inhomogeneous contrast medium uptake due to central areas of necrosis; i: Typical myelolipoma.

 

Magnetic Resonance Imaging (MRI)

 

Adrenal imaging with MRI can also aid in the differential diagnosis between benign and malignant adrenal pathology. Benign adrenal adenomas appear hypotense or isotense compared to the liver on T1-weighted images and have low signal intensity on T2-weighted images. The majority of PCCs show high signal intensity on T2-weighted imaging (“light bulb sign”) which is a non-specific finding; however, a wide range of imaging features of PCCs mimicking both benign and malignant adrenal lesions have also been described (79). Primary ACCs are characterized by intermediate to high signal intensity on T1- and T2-weighted images and heterogeneity (mainly on T2- sequence due to hemorrhage and/or necrosis) as well as avid enhancement with delayed washout. However, these features are not specific and display significant overlap between benign and malignant lesions. The MRI technique of chemical-shift imaging (CSI) exploits the different resonance frequencies of protons of water and triglyceride molecules oscillating in- or out-of-phase to each other under the effect of specific magnetic field sequences, to identify high lipid content in adrenal lesions (86). Adrenal adenomas with a high content of intracellular lipids usually lose signal intensity in out-of-phase images compared to in-phase images, whereas lipid-poor adrenal adenomas, malignant lesions, and PCCs remain unchanged. Signal intensity loss can be assessed qualitatively by simple visual comparison or by quantitative analysis using the adrenal-to-spleen signal ratio and can identify adenomas with a sensitivity of 84-100% and a specificity of 92-100% (87). It must be noted however, that ACC and clear renal cell cancer metastases may sometimes also show signal loss (88).

 

The evidence regarding the diagnostic accuracy of MRI is generally considered poor for several reasons, such as: low number and quality of studies, lack of standardized quantitative assessment, subjective interpretation of qualitative loss in signal intensity, and paucity of recent high-quality research. Additionally, there are no good quality studies comparing the diagnostic performance of MRI and CT in AIs. Hence, based on the higher strength of evidence, CT is considered the primary radiological procedure for evaluating AIs, being also more easily available and cost-effective. MRI should be reserved for cases in which CT is less desirable (as in pregnant women and in children) (66,89).

 

Figure 3. MRI images of different adrenal lesions presenting as incidentalomas, using the chemical shift imaging (CSI) technique. The loss of signal in out of phase images is typical in benign lipid-rich adenomas (a, b) in contrast with pheochromocytomas (c, d) and adrenocortical carcinomas (e, f) which do not display any signal loss.

 

Scintigraphy

 

In recent years, positron emission tomography (PET) using 18-fluoro-deoxyglucose (18F-FDG) has emerged as an effective tool in identifying malignant adrenal lesions. By utilizing the increased glucose uptake properties of cancer cells, 18F-FDG-PET combined with a CT scan (18F-FDG-PET/CT) achieves a sensitivity and specificity in identifying malignancy of 93-100% and 80-100% respectively (90,91). Both quantitative analysis of FDG uptake using maximum standardized uptake values (SUVmax) and qualitative assessment using a mass/liver SUV ratio have been used as a criterion, with the latter displaying better performance (92). A SUV ratio <1.45–1.6 between the adrenal and the liver is highly predictive of a benign lesion (93). Caveats in utilizing 18F-FDG-PET/CT include cost and availability, risk of false negative results in the case of necrotic or hemorrhagic malignant lesions, size <1cm, extra-adrenal malignancies with low uptake (such as metastases from renal cell cancer or low-grade lymphoma), and false positive results in cases of sarcoidosis, tuberculosis, and other inflammatory or infiltrative lesions and some adrenal adenomas and PCCs that show moderate FDG uptake (94). Because of its excellent negative predictive value, 18F-FDG-PET may help in avoiding unnecessary surgery in patients with non-secreting tumors with equivocal features in CT demonstrating low FDG uptake. Moreover, 18F-FDG-PET/CT may favor surgical removal of tumors with elevated uptake and no biochemical evidence of a PCC (90). Newer PET tracers such as 18F-fluorodihydroxyphenylalanine (F-DOPA) and 18F-fluorodopamine (FDA) for detection of PCC have also been developed but their availability is limited (95).

Conventional adrenal scintigraphy using radiolabeled cholesterol molecules such as 131I-6-b-iodomethyl-norcholesterol (NP-59) and 75Se-selenomethyl-19-norcholesterol has been used in the past to discriminate benign from malignant lesions. These tracers enter adrenal hormone synthetic pathways and act as precursor-like compounds, providing information regarding the function of target tissue. Typically, benign hypersecreting tumors, and non-secreting adenomas, show tracer uptake, whereas primary and secondary adrenal malignancies, space-occupying or infiltrative etiologies of AIs appear as ‘cold’ masses, providing an overall sensitivity of 71-100% and a specificity of 50-100% (96). However, some benign adrenal tumors such as myelolipomas and some functioning ACCs, may also be visualized with these modalities. Several additional limitations of adrenal scintigraphy such as insufficient spatial resolution, lack of widespread expertise, limited availability of the tracer, being a time-consuming procedure (which requires serial scanning over 5-7 days), and high radiation doses received by the patient, have limited its value in routine clinical practice, especially when conventional imaging can provide more reliable information. Recently, 123I-iodometomidate has been introduced as a tracer because it binds specifically to adrenocortical enzymes, but its application is hampered by its limited availability and heterogeneous uptake by ACCs (97). Scintigraphy with 123I-meta-iodo-benzyl-guanidine (MIBG) is the preferred method for identifying PCCs when clinical, biochemical, and imaging features are not conclusive, or when multiple or malignant lesions need to be excluded (40).

 

Table 2 summarizes the imaging properties of different underlying AI pathologies that can be helpful for the differential diagnosis.

 

Table 2. Image Findings Differentiating Common Adrenal Pathologies in AIs

FINDING

Benign adenoma

ACC

Pheochromocytoma

Metastases

Size

Usually <4cm

Usually >4cm

Variable

Variable

Growth rate

Stable or <0.8cm/year

Significant growth (>1cm/year)

Slow growth

Significant growth (>1cm/year)

Shape & margins

Round or oval with well-defined margins

Irregular shape and margins. Invasion to surrounding tissues

Variable

Variable

Composition

Homogenous

Heterogeneous (hemorrhage, necrosis)

Heterogeneous (necrosis)

Heterogeneous (hemorrhage, necrosis)

CT Unenhanced attenuation

≤10 HU (or >10 HU for lipid-poor adenomas)

>10 HU

>10 HU

>10 HU

CT Percent Washout (PW)

APW >60%

RPW>40%

APW<60%, RPW<40%

APW<60%

RPW<40%

APW<60%, RPW<40%

MRI – CSI

(out-of phase)

Signal loss

(except in lipid-poor adenomas)

No change in signal intensity

No change in signal intensity

No change in signal intensity

FDG uptake (PET)

Low (some can have low to moderate uptake)

High

Low (malignant pheochromocytomas show high uptake)

High

NP-59 uptake

Present

Absent (except in some secreting tumors)

Absent

Absent

ACC: Adrenocortical carcinoma; HU: Hounsfield Units; APW: Absolute PW; RPW: Relative PW; CSI: Chemical-shift Imaging; FDG: fluoro-deoxyglucose; NP-59: 131I-6-b-iodomethyl-norcholesterol

 

HORMONAL EVALUATION

 

Patients with AIs should be screened at presentation for evidence of excess catecholamine or cortisol secretion and, if hypertensive and/or hypokalemic, for aldosterone excess. As already discussed, the definition of AI per se implies the absence of clinical symptoms/signs related to these entities, however subtle hormonal hypersecretion not leading to the full clinical phenotype of a related syndrome may be present in patients with an AI (6).

 

Screening for Cortisol Excess

 

According to the Endocrine Society’s Clinical Practice Guidelines for the diagnosis of Cushing’s syndrome and the AACE/AAES Medical Guidelines for the management of AIs, all patients with an incidentally discovered adrenal mass should be tested for the presence of hypercortisolism (64,98). Signs and symptoms of overt Cushing’s syndrome if present in a thorough clinical evaluation should prompt the physician to proceed with the recommended diagnostic approach described in the relevant Endocrine Society’s Clinical Guidelines (98). In this case, as discussed earlier, the validity of the term “incidentaloma” is debated.

 

In the absence of overt disease, biochemical investigation frequently reveals subtle cortisol hypersecretion and abnormalities of the HPA axis, a state previously termed as subclinical Cushing’s syndrome (6). Based on the most recent clinical practice guidelines by the European Society of Endocrinology (ESE) and European Network for the Study of Adrenal Tumors (ENSAT) the term “mild autonomous cortisol secretion” (MACS) is preferred and will also be used throughout this chapter. Although MACS is poorly defined, and its natural history is unclear (3), the prevalence of hypertension, diabetes, obesity, other features of the metabolic syndrome, and osteoporosis has been found to be increased in such patients (5,99). Because standard biochemical tests used to screen for Cushing’s syndrome were not designed to reveal the subtle changes encountered in MACS, and since a definitive clinical phenotype to ascertain the presence of this condition is missing, a combination of various parameters used to assess the integrity of the HPA axis have been employed. Alterations of the HPA axis suggestive of MACS in AIs include altered dexamethasone suppression (DST) and response to CRH, increased mean serum cortisol and urinary free cortisol (UFC) levels, reduced dehydroepiandrosterone sulfate (DHEA-S) and reduced ACTH levels (33), although the latter has recently been questioned since most ACTH assays lack sensitivity at the lower part of the reference range (100). Incorporation of midnight salivary cortisol as a means to diagnose MACS has produced inconsistent results (101).

 

Currently, the 1 mg overnight DST, remains the most reliable and easily reproducible method and is the recommended test to detect cortisol secretion abnormalities based on pathophysiological reasoning, simplicity, and incorporation in the diagnostic algorithms of most studies. (5,101). Cortisol autonomy in AIs reflects a biological continuum without a clear separation between functioning and non-functioning tumors. Different cortisol cut-off values following the 1 mg DST have been advocated from different authors and were adopted by several authorities, ranging from 50 to 138 nmol/l (1.8 to 5 μg/dl) (64,102). Higher thresholds increase the specificity of the test but lower its sensitivity (103). The post 1 mg DST cortisol cutoff of >5 μg/dl (138 nmol/l) approach was substantiated by studies showing that all patients with such a cortisol value had uptake only on the side of the adenoma on adrenal scintigraphy (104). On the other hand, studies that used post-surgical hypoadrenalism as indicative of autonomous cortisol secretion suggested that lower cortisol cut-offs may be needed to identify these cases (105–107). Furthermore, older stratification of autonomy based on different post-1mg ODST cortisol levels has been abandoned by recent guidelines (66). A negative DST using a cortisol cut-off value of 1.8 μg/dl (50 nmol/l) virtually excludes MACS. Furthermore, several studies have found that patients with post DST cortisol values >1.8 μg/dl (50 nmol/l) have increased morbidity or mortality (108,109) .The formal low dose dexamethasone suppression test (LDDST) can be used to confirm and quantify the degree of autonomous cortisol secretion or to exclude a false positive test (110,111). Based on our experience, the post-LDDST cortisol value should be considered in patients with such intermediate cortisol values following the 1 mg DST because, in addition to its high specificity, it correlates well with other indices of cortisol excess and the size of the adenoma, thus providing a quantitative measure of the degree of cortisol production from the adenoma and a more robust means for further follow-up (110,112). Although confirmation of ACTH independency (through suppressed ACTH levels) is also required to establish the diagnosis of MACS (64), the 1 mg DST should be the initial screening test based on pathophysiology and the fact that it represents the most common HPA axis abnormality reported by most studies (49). It should also be noted that cortisol levels after 1mg DST are increasing with age, making the diagnosis of MACS in frail elderly patients difficult. Especially for this subgroup of patients in which comorbidities are already frequently present, MACS diagnosis is not considered clinically relevant, and could be omitted. Finally, it is important to consider drugs or conditions that interfere with this test by altering dexamethasone absorption, metabolism by CYP3A4, or falsely elevate cortisol levels through increased cortisol-binding globulin (CBG) levels (113). Consequently, repeating the 1mg overnight DST in patients who were previously tested positive, and especially those who are candidates for surgery, is advisable.

 

Reduced levels of DHEA-S also reflect chronic suppression of ACTH secretion and have been found to offer comparable sensitivity and greater specificity to the existing gold-standard 1 mg DST for the diagnosis of MACS (114). In a study of 185 patients with AIs of which 29 patients (16%) were diagnosed with autonomous cortisol secretion, an age- and sex-specific DHEA-S ratio (derived by dividing the DHEA-S by the lower limit of the respective reference range for age and sex) of <1.12 was >99% sensitive and 92% specific for the diagnosis of MACS (115). In a retrospective study of 256 patients with AIs and MACS, a serum DHEA-S concentration <40 μg/dL was 84% specific for MACS, whereas an ACTH concentration <10 pg/mL was only 75% specific for MACS. In addition, a serum concentration of DHEAS >100 μg/dL combined with an ACTH >15 pg/mL was 96% percent specific for excluding MACS (116). The only caveat is that age- and sex- adjusted DHEA-S reference values are not well established.

 

Recently, studies utilizing gas chromatography-tandem mass spectrometry (GC-MS/MS) to measure serum and 24-hour urine levels of several steroids in patients with AIs have emerged, showing promising potential. Patients with MACS have been found to have decreased levels of adrenal androgens and their metabolites and increased levels of glucocorticoid metabolites compared to healthy individuals, with sensitivity and specificity rates comparable to routine methods (117–119).

 

Since cortisol-related comorbidities play such an important role in planning patient management, it is crucial to gather medical information and laboratory data about glucose and lipid metabolism, hypertension, bone density and fractures. 

 

Screening for Pheochromocytoma

 

Although arterial hypertension and other signs of catecholamine excess are considered classical clinical manifestations of PCCs, screening should be performed even in normotensive patients with AIs since catecholamine secretion can be intermittent, and cases of “silent” PCCs are increasingly being recognized (120). The initial recommended biochemical screening test is measurement of plasma free (from blood drawn in the supine position) or urinary fractionated metanephrines using liquid chromatography with mass spectrometric or electrochemical detection methods (40). This approach has a sensitivity and specificity of 99% and 97% respectively and has proven to be superior to measurement of plasma or urine catecholamines and vanillylmandelic acid (VMA) (121). The   issue   concerning   the   diagnostic   performance   of   plasma   free   versus   urinary   fractionated metanephrines has been recently settled in a multicenter prospective study involving over 2,000 patients, with follow-up to exclude patients without PPGL and with LC-MS/MS measurements of plasma and urinary free metanephrines compared to urinary deconjugated metanephrines (122). In this study, diagnosis of PPGLs using plasma or urinary free metabolites provided advantages of fewer false-positive results compared with commonly measured de-conjugated metabolites. The plasma panel offered better diagnostic performance than either urinary panel for high-risk patients but was comparable for patients at low risk of disease. Sane et al suggested that routine biochemical screening for PCC in small (<2cm) homogenous AIs characterized by attenuation values <10 HU may not be necessary, since none of the 115 patients in his cohort with lipid-rich tumors (<10 HU) had constantly elevated 24-hour urinary metanephrines or normetanephrines, whereas all 10 histologically proven PCCs were larger than 2cm and were characterized by >10 HU in unenhanced CT scans (123). This was also confirmed from a recent multicenter retrospective study including 376 PCCs with sufficient data from CT imaging. Based on the lack of PCCs with an unenhanced attenuation of <10 HU and the low proportion (0.5%, 2/376) of PCCs with an attenuation of 10 HU, it was suggested that abstaining from biochemical testing for PCC in AIs with an unenhanced attenuation of ≤10 HU is reasonable, whereas contrast washout measurements were unreliable for ruling out PCC (124).

 

A recent study (125) comparing the clinical, hormonal, histological, and molecular features of normotensive incidentally discovered PCCs (previously referred as “silent”) with tumors causing overt symptoms, revealed lower diagnostic sensitivity (75%) for plasma and urinary metanephrines irrespective of tumor size, while genetic and histological studies showed decreased expression of genes and proteins associated with catecholamine production and increased cellularity and mitotic activity in “silent” tumors. It was implied that asymptomatic incidentally discovered PCCs do not represent an early stage of development of PCCs but rather correspond to a distinct entity characterized by cellular defects in chromaffin machinery resulting in lower efficiency to produce or release catecholamines. It is, therefore, crucial to consider that normotensive patients with an AI and normal values of metanephrines, may indeed harbor a PCC. In such instance, the CT and MRI scan features of the tumor if suspicious for PCC, should alert the clinician to perform complementary investigations, such as plasma chromogranin A measurement, MIBG scintigraphy, 18F-FDG-PET/CT, or other alternative functional imaging (F-DOPA/PET or FDA/PET) to rule out this possibility.

 

Screening for Aldosterone Excess

 

According to published guidelines from the Endocrine Society, all patients with an AI and hypertension, irrespective of serum potassium levels, should be tested for PA using the plasma aldosterone/renin ratio (ARR) as a screening test (42). However, the knowledge that PA can be diagnosed in normotensive patients with hypokalemia necessitates testing of all patients with hypertension or hypokalemia (44). Although there is no current consensus regarding the most diagnostic ARR cut-off, values >20-40 (plasma aldosterone expressed as ng/dl and plasma renin activity [PRA] as ng/ml/h) obtained in the morning from a seated patient are highly suggestive. However, the plasma aldosterone level also needs to be considered because extremely low PRA, even in the presence of normal aldosterone levels, will result in a high ARR; an aldosterone level less than 9 ng/dl makes the diagnosis of PA unlikely, whereas a level in excess of 15 ng/dl is suggestive (49). Attention should also be given in certain technical aspects required for the prompt interpretation of the ARR such as unrestricted dietary salt intake, corrected potassium levels, and washout of interfering antihypertensive medication. Patients may be treated with a non-dihydropyridine calcium channel blocker (verapamil slow release) as a single agent or in combination with α-adrenergic blockers (such as doxazosin) and hydralazine for blood pressure control during the washout period, if needed.

 

When suspected based on the ARR, PA should be verified with one of the commonly used confirmatory tests (oral sodium loading, saline infusion, fludrocortisone suppression, and captopril challenge). Admittedly, the extent that patients with AI should be investigated to exclude PA is still not known. Although PA has been reported with a low prevalence between patients with AIs (1-10%), substantially higher rates (24%) have recently been described using a recumbent post-low dose dexamethasone suppression (LDDST)-saline infusion test (PD-SIT) (45). Further studies evaluating the optimal biochemical diagnostic approach of PA in patients with AIs are required by comparing established versus evolving investigational protocols.

 

Screening for Androgen/Estrogen Excess

 

Measurement of sex hormones is not recommended in patients with an AI on a routine basis (64). Elevated levels of serum DHEA-S, androstenedione, 17-OH progesterone as well as testosterone in women and estradiol in men and postmenopausal women can be found in more than half of patients with ACCs (126). Although cases of androgen or estrogen excess have been rarely described in patients with benign adrenocortical adenomas (127–130), they are usually accompanied by symptoms or signs of virilization in women (acne, hirsutism) or feminization in men (gynecomastia), and therefore such lesions cannot be considered as true AIs. Thus, the usefulness of measuring sex hormones and steroid precursors is limited to cases of adrenal lesions with indeterminate or suspicious for malignancy imaging characteristics, where elevated levels can point towards the adrenocortical origin of the tumor and suggest the presence of an ACC rather than a metastatic lesion. Additionally, increased basal or after cosyntropin stimulation levels of 17-OH progesterone can also indicate CAH in patients with bilateral AIs (6).

 

Screening for Hypoadrenalism

 

Bilateral AIs caused by metastases of extra-adrenal malignancies or infiltrative diseases can rarely cause adrenal insufficiency (131). Therefore, in all patients with bilateral adrenal masses, adrenal insufficiency should be considered and evaluated clinically and if likely, diagnosis should be established using the standard 250μg cosyntropin stimulation test according to the Endocrine Society’s recently published clinical guidelines (132). 

 

FINE NEEDLE ASPIRATION BIOPSY (FNAB)

 

Percutaneous fine-needle aspiration biopsy (FNAB) as means to clarify the nature of an AI has now been surpassed by the non-invasive radiological methods because they have better diagnostic accuracy and are devoid of potential side effects (133,134). It should be noted that FNAB is not considered an accurate method in differentiating benign from malignant primary adrenal tumors (135) but can be helpful in the diagnosis of metastases from extra-adrenal malignancies, lymphoma, sarcoma, infiltrative or infectious process with a sensitivity of 73-100% and a specificity of 86-100% using variable population inclusion criteria, reference standards, and biopsy techniques (136–138). Adrenal biopsy is not needed if the patient is already known to have widespread metastatic disease. Biopsy is only recommended for hormonally inactive masses not characterized as benign on imaging and where a biopsy result would affect treatment decisions. FNAB has significant procedural risk with complications such as pneumothorax, bleeding, infection, pancreatitis, and dissemination of tumor cells along the needle track reported at a rate up to 14% by some, but not all available studies (133). To avoid the risk of a potentially lethal hypertensive crisis, PCC should always be excluded biochemically before FNA of an adrenal mass is attempted (139).

 

NATURAL HISTORY OF AIs

 

Since AIs do not represent a single clinical entity, their natural history varies depending on the underlying etiology. Primary malignant adrenal tumors typically display rapid growth (>2 cm/year) and a poor outcome with an overall 5-year survival of 47%. It is not known whether prognosis of patients with incidentally discovered ACC is different from symptomatic cases, however detection of the tumor at an early stage provides the possibility of definitive surgical cure (140). Patients bearing adrenal metastases have a clinical course depending on stage, grade, and site of the primary tumor (4). PCCs grow slowly and are mostly benign, but if untreated are potentially lethal displaying high cardiovascular mortality and morbidity, whereas 10-17% of the cases can be malignant (40). This is further emphasized by the fact that PCCs detected in autopsy series had not been suspected in 75% of the patients while they were alive, although they contributed to their death in approximately 55% of cases (141).

 

In benign adrenal tumors, which constitute the majority of AIs, the main concerns about their natural history revolve around their progressive growth, the possibility of malignant transformation, and the risk of evolution towards overt hypersecretion. Several cohort studies, despite their limitations, have shown that the majority of benign tumors remain stable in size; only 5-20% show a >1 cm increase in size, mostly within the first three years after prolonged follow-up (142,143), whereas occasional shrinkage, or even complete disappearance, of an adrenal mass have also been reported in about 4% of cases (8,144). Although there is not a specific growth rate cut-off indicative of a benign nature, ACCs initially presenting as AIs, are invariably characterized by a rapid growth within months (at least > 0.8cm/year). The risk of an AI initially considered to be benign to become malignant has been estimated at <1/1000 (3,8) by Cawood et al, who found only two reports of a malignancy detected during the follow-up of AIs presenting as benign at diagnosis; the first was a renal carcinoma metastasis in a patient with a known history of renal carcinoma and the other was a non-Hodgkin’s lymphoma that showed a mass enlargement after 6 months (3). Two case reports of patients with a well-documented history of adrenal incidentalomas with totally benign imaging features on CT, who were diagnosed on follow-up (8 and 14 years later) with a malignant tumor in the same adrenal gland have recently been described (145,146). It is not known whether these cases can be explained by the independent occurrence of two events in a single adrenal (initially a typical benign adenoma and consequently the occurrence of an ACC) or whether a malignant transformation of a benign adenoma to carcinoma was the underlying course of events. Although there is evidence to suggest the adenoma-carcinoma sequence is possible in the adrenal cortex (147,148), the high prevalence of adenomas contrasting with the extremely low prevalence of ACCs suggest that this process is probably exceptionally rare. These findings highlight the low risk of malignant transformation of AIs and the adequacy of current imaging to ascertain the diagnosis at presentation deterring the need for long-term imaging follow-up.

 

The appearance of hormonal hypersecretion over time in initially NFAIs varies in different series. New-onset catecholamine or aldosterone overproduction is extremely rare (<0.3%), whereas development of overt hypercortisolism during follow-up is found in <1% (8). The most common disorder observed during follow-up is the occurrence of autonomous cortisol secretion eventually leading to MACS, reported with a frequency of 5.4% (CI 3,1-8,1%) (66,144). This risk is higher for lesions >3 cm in size and during the first 2 years of follow-up but seems to plateau after 3-4 years, even if it does not subside completely (149). On the other hand, subtle hormonal alterations discovered at initial screening may also improve over time, indicating possible cyclical cortisol secretion from AIs and/or highlighting the inherent difficulty in biochemical confirmation of this condition (143).

 

Another issue of debate regarding the natural history of AIs that has attracted research, producing frequently conflicting data, is the sequelae of MACS on cardiovascular risk and subsequent mortality and morbidity. Several cross-sectional and cohort studies have reported a clustering of unfavorable cardiovascular risk factors in patients with AIs similar to those found in patients with overt Cushing’s syndrome (150,151). It is biologically plausible to anticipate that the presence of even mild to minimal cortisol excess may lead to some extent to the classic long-term consequences of overt hypercortisolism, such as hypertension, obesity, impaired glucose tolerance or frank diabetes, dyslipidemia, and osteoporosis (figure 4). Because these metabolic derangements are common in the general and particularly the elderly population, in whom AIs are more frequently found, it is difficult to extrapolate whether there is a causal relationship between them. Whether these metabolic abnormalities in patients with AIs result in increased cardiovascular mortality and morbidity has not as yet been fully clarified. Although, some recent retrospective studies (108,109,152,153) have shown higher rates of cardiovascular events and mortality in patients with higher cortisol levels after the 1 mg DST, data from patients who underwent adrenalectomy are contradictory, regarding the outcome on metabolic and cardiovascular profile, whereas there are relatively few data on the risk of major cardiovascular events or mortality (107,154–156). Similarly, evidence on the detrimental effects of MACS on bone metabolism, such as lower bone density and high prevalence of vertebral fractures (43-72%) in postmenopausal women and eugonadal male patients with AIs (99,157–160) are conflicting with studies not showing reversal of these effects following surgical treatment (154,161). Additionally, most of the detected vertebral fractures were minor and of uncertain clinical impact (99).

 

Moreover, there is growing evidence that even non-functioning Ais (NFAIs) may be associated with similar metabolic disturbances and manifestations of the metabolic syndrome that are considered cardiovascular risk factors (162–164). Compared with controls, patients with NFAIs exhibit subtle indices of atherosclerosis such as increased carotid intima-media thickness (IMT)(165), impaired flow-mediated vasodilatation (FMD) (166), and left ventricular hypertrophy (167). A recent study excluding patients with traditional risk factors (diabetes, hypertension or dyslipidemia) reported similar findings in patients harboring NFAIs, with increased insulin resistance and endothelial dysfunction that correlated with subtle but not autonomous cortisol excess (168). Furthermore, an observational study suggested that patients with NFAIs had a significantly higher risk of developing diabetes compared with control subjects without adrenal tumors prompting a re-assessment of whether the classification of benign adrenal tumors as “non-functional” adequately reflects the continuum of hormone secretion and metabolic risk they may harbor (169).

 

A recent meta-analysis (170) of 32 studies including patients with NFAIs and adrenal tumors associated with MACS provided important insights on the natural history of such tumors that help in solving controversy and informing practice. First and foremost, it was observed that only a small proportion of patients with NFAI or MACS had tumor growth or changes in hormone production during follow-up. Only 2.5% of adrenal incidentalomas grew by 10 mm or more over a mean follow-up of 41.5 months, whereas the mean difference in adenoma size between follow-up and baseline in all patients was negligible at 2.0 mm. Larger adenomas at diagnosis (≥25 mm) were even less likely than smaller tumors to grow during follow-up, which, according to the authors, suggests attainment of maximum growth potential. More importantly malignant transformation was never observed at the end of follow-up. Similarly, in patients with NFAIs or MACS at diagnosis, the risk of developing clinically overt hormonal hypersecretion syndromes (Cushing’s, PA, or catecholamine excess) was negligible (<0,1%), suggesting that these rare cases are probably attributed to the development of subsequent adrenal tumors and that MACS does not represent a preliminary stage of overt Cushing’s. Inapparent cortisol autonomy ensued only in 4.3% of patients with initially nonfunctioning tumors. The third and most novel finding of this thorough meta-analysis pertained to comorbidities, cardiovascular risk, and mortality. It was confirmed, like in other similar studies, that patients with MACS had a high prevalence of cardiovascular risk factors (such as hypertension, obesity, dyslipidemia, and type 2 diabetes) and were more likely than those with NFAIs to develop or show worsening of these factors during follow-up. However, the prevalence of such factors in patients with NFAIs was also significant and higher than expected for Western populations. This finding could be explained by a subtle degree of glucocorticoid excess not detected by current diagnostic criteria or perhaps by cyclical cortisol secretion or even by excess cortisol secretion in response to stress situations. It could also represent ascertainment bias since patients with diseases are more likely to have imaging tests that may detect an AI or could be a result of the previously theorized reverse causality concept that diabetes or the metabolic syndrome promote adrenal tumor development (171). Interestingly, reported all-cause and cardiovascular mortality in patients with NFAI during follow-up were similar to those in patients with MACS, warranting close clinical follow-up and treatment for both groups of patients.

 

MANAGEMENT

 

A proposed algorithm for diagnostic approach and management of AIs based on the more recently published and widely accepted guidelines (66) is presented in Figures 4 and 5. A patient presenting with a newly discovered AI should be initially assessed in parallel for its malignancy potential and functional status. Exclusion of malignancy is critical and imaging review by an experienced radiologist is of crucial importance. Since evidence for the accuracy of MRI-CSI is not as strong, non-contrast CT is the first modality that should be used if not already performed. An unenhanced attenuation value of ≤ 10 HU combined with homogeneity can safely, based on available data, confirm the diagnosis of benign adenoma and exclude malignancy, requiring no further imaging investigation or follow-up. The same can be applied for larger AIs (>4cm) with unequivocal benign phenotype (≤ 10 HU, homogeneous), since recent observational data have provided better quality evidence for their benign natural course (72,172). For tumors with >10 HU, management is dependent on the risk of malignancy based on a combination of imaging properties such as attenuation (11-20 HU or >20 HU), size (< or > 4cm) and homogeneity (homogeneous or heterogeneous). In a homogeneous, < 4 cm adrenal mass with unenhanced HU between 11 and 20, the likelihood of malignancy is <10%. Thus, the proposed approach is to immediately acquire an additional imaging study, depending on the local experience and preference (FDG-PET/CT, MRI with CSI or CT with washout protocol). If the findings from the additional imaging are suggestive of a benign lesion, no further imaging follow-up is required. Alternatively, interval imaging (with non-contrast CT or MRI) after 12 months could be performed, to ensure that no significant change in size has occurred. On the opposite side, AIs that have relatively high risk of malignancy should be discussed in a multidisciplinary team (MDT) meeting. Those include AIs ≥4 cm with density > 20 HU or a heterogeneous appearance and are most likely candidates for immediate surgical removal. Prior to surgery staging with chest CT and/or FDG/PET-CT is recommended to detect metastatic disease if present. In case the MDT recommendation is not surgery, interval imaging (with non-contrast CT or MRI) in 6-12 months is advised. All other AIs with intermediate tumor characteristics (tumor size ≥ 4 cm with unenhanced HU 11-20, or tumor size < 4 cm with unenhanced HU > 20, or tumor size < 4 cm with heterogeneous appearance), have a smaller but considerable relative risk for malignancy and should be examined in detail in an MDT meeting. Ordering additional imaging (FDG-PET/CT, MRI with CSI or CT with washout protocol, depending on local availability and expertise) seems to be the appropriate strategy. In these cases, additional imaging with FDG/PET-CT might have an advantage over the other modalities due to the low risk of false negative results. If the tumor remains indeterminate after the additional imaging workup, surgery or interval imaging (with non-contrast CT or MRI) after 6-12 months could be offered. A promising alternative to additional imaging, that has appeared in recent years, is urine or plasma steroid metabolomics (profiling) by tandem mass spectrometry. In two published retrospective studies (72,119), one using urine and the other plasma samples, sensitivity for excluding adrenocortical cancer, as stand-alone tests, was approximately 80%. However, when combined with imaging properties (namely attenuation >20 HU and size >4cm) urine steroid metabolomics showed a negative predictive value of 99.7%.

 

Interval imaging at 6 and/or 12 months in case no surgery is performed (MDT decision or for any other reason) is done to monitor possible progressive growth. An increase of >20% of the largest tumor diameter together with an at least 5 mm increase in this diameter (101), as defined by RECIST 1.1 criteria, or an absolute increase by >8 mm over 12 months, as suggested by some studies (67), probably warrant re-evaluation by the MDT. Further imaging follow-up may not be needed if no change is size is seen at the first interval imaging.

 

In indeterminate cases, age is a parameter that needs to be considered by the MDT when deciding which patients to refer for adrenalectomy. For example, most clinicians would tend to advise in favor of removing a lipid-poor (19 HU) 3.2 cm AI in a 23-year-old woman, whereas serial imaging follow-up would be favorably recommended in an 83-year-old woman with a lipid-poor (15 HU) 4.7 cm adrenal tumor.

 

All published guidelines and expert reviews agree that patients with unilateral adrenal masses causing unambiguous hormonal overactivity, and those with suspected malignancy (mainly ACC), are candidates for surgical interventions (5,6,40,42,64,66,101,102,173,174). There is also broad consensus that the majority of AIs with clearly benign imaging phenotype in unenhanced CT and no evidence of functionality do not require surgery.

 

The management of patients harboring AIs who have MACS is debatable and the beneficial effect of adrenalectomy has not been proven adequately in the literature. Some, but not all, predominantly retrospective studies have shown a beneficial effect in hypertension and diabetes mellitus in patients with AIs who underwent an adrenalectomy, compared to those who did not undergo such a procedure (107,154,156). In one prospective study with an 8-year follow-up, operated patients with MACS had an improvement in features of the metabolic syndrome, but not of osteoporosis, compared to those who were conservatively managed; however, no control group was included in the study (154). An improvement of blood pressure and blood glucose was noted in a retrospective study of adrenalectomized patients with MACS, whereas these indices worsened in non-operated patients; even so, some patients apparently with NFAI also showed an improvement in some of these parameters (107). In a recent prospective multicenter randomized study including 62 patients aged 40-75, Morelli et al showed that adrenalectomy more frequently ameliorated hypertension (68% versus 13%) and glycometabolic control (28% versus 3,3%) than the conservative approach, while the latter was associated with a more frequent worsening of blood pressure and insulin resistance (12% versus 40%). Since available data from the aforementioned retrospective and the two recent small prospective studies are not considered high-quality, the decision to recommend surgery should be taken in a multidisciplinary setting while taking several other factors into consideration, such as: duration and evolution of comorbidities and their degree of control, presence and extent of end organ damage inappropriate for age, discrepant family history, presence of multiple comorbidities, age, sex, general health, degree and persistence of nonsuppressible cortisol after dexamethasone, and patient’s preference. Young patients with MACS and those with new onset and/or rapidly worsening comorbidities resistant to medical treatment(6,175) could thus be candidates for surgical intervention.

 

Myelolipomas are considered benign tumors, their diagnosis is mostly based on imaging characteristics and biochemical evaluation is not usually needed unless informed by clinical presentation. Measurement of 17(OH) progesterone is advised in large and/or bilateral myelolipomas for the possibility of CAH. Their management is mostly conservative with yearly imaging follow-up, since in up to 16% of the cases a median tumor growth of at least 1cm per year was demonstrated. Surgery is usually reserved for large tumors, those with tumor growth, acute hemorrhage, symptoms of abdominal mass effect, or uncontrolled CAH (176).

 

Before proceeding to surgical therapy, appropriate medical therapy must be given to all functioning lesions, aiming at symptom control. Apart from patients with Cushing’s syndrome, post-surgical adrenal insufficiency may ensue in MACS patients (177,178). Because the need for glucocorticoid coverage cannot be predicted before surgery, patients should be covered by steroids post-operatively until the HPA-axis can be formally assessed (105). Low morning cortisol levels the day after surgery, and before glucocorticoid replacement, provide evidence for post-surgical hypoadrenalism (107). All patients diagnosed with PCC, including normotensive patients with “silent” tumors should receive preoperative α-adrenergic blockade for 7 to 14 days to prevent perioperative cardiovascular complications. Treatment should also include a high-sodium diet and fluid intake to reverse catecholamine-induced blood volume contraction preoperatively and prevent severe hypotension after tumor removal (40). Finally, patients diagnosed with PA and bilateral tumors or a unilateral AI (if older than 40 years of age) who seek a potential surgical cure, should be considered for adrenal venous sampling (AVS) before proceeding to surgery, to confirm lateralization of the source of the excessive aldosterone secretion. In cases where decision for adrenalectomy is based on imaging phenotype it would also be prudent to exclude the possibility of a “silent” PCC before proceeding to surgery, because hemodynamic instability during surgical excision may ensue.

 

According to earlier published AACE/AAES Medical Guidelines for the management of adrenal incidentalomas, patients with AIs not elected for surgery after the initial diagnostic work-up, should undergo re-imaging 3-6 months after the initial diagnosis and then annually for the next 1-2 years, while annual biochemical testing is advised for up to 4-5 years following the diagnosis (64). However, it has recently been suggested by some authors that given the low probability of the transformation of a benign and non-functioning adrenal mass to a malignant or functioning one, the routine application of the current strategies in all patients with AIs is likely to result in a number of unnecessary biochemical and radiological investigations (3,179,180). Such an approach is costly, and it does not take into account harmful consequences of diagnostic evaluation such as patients’ anxiety associated with repeated clinical visits and a high rate of false positive results leading to further testing or unnecessary adrenalectomy. Moreover, exposure to ionizing radiation from repeated CT scans increases the future cancer risk to the level that is similar to the risk of the adrenal lesion becoming malignant (3,181).

 

Patients without any biochemical abnormalities at presentation could be spared the burden of repeated testing, since the risk of developing clinically overt hormonal excess is extremely low. Clinical follow-up with assessment of cardiovascular risk factors that have been associated with the presence of AIs may be adequate to detect the reported ~10% of the cases of new onset MACS (5). Patients with worsening of their metabolic parameters should be retested with the 1mg DST and be advised to apply lifestyle changes and effective medical treatment to reduce cardiovascular risk. If biochemical abnormalities suggesting MACS are present during the initial screening, annual clinical follow-up including evaluation of potentially cortisol excess-related comorbidities, as well as periodic testing of the HPA axis, is advisable. Patients with MACS who do not reach the treatment goals despite an adequate medical therapy could be offered surgery. Duration of follow-up is also under debate, however based on available data, annual hormonal evaluation may be suggested for up to five years, and especially for lesions >3 cm (64).

 

CONCLUSION

 

AIs are increasingly being recognized, particularly in the aging population. Adrenal CT and MRI can reliably distinguish benign lesions, while 18F-FDG-PET/CT scan can be helpful in identifying tumors with malignant potential. MACS is the most common hyperfunctional state that is best substantiated using the 1 mg DST; urinary/plasma metanephrines and ARR are used to screen for PCCs and hyperaldosteronism. Adrenal lesions with suspicious radiological findings, PCCs and tumors causing overt clinical syndromes, as well as those with considerable growth during follow-up, should be treated with surgical resection. Although there is no consensus, the interval for diagnostic follow-up testing relies on the radiological and hormonal features of the tumors at presentation. The benefit of surgical resection in patients with substantial comorbidities and associated subclinical adrenal hyperfunction, mainly in the form of MACS, is still under investigation.

 

Figure 4. Proposed algorithm for diagnosis and management of AIs (imaging evaluation).

Figure 5. Proposed algorithm for diagnosis and management of AIs (biochemical evaluation)

 

REFERENCES

 

  1. Korobkin M, White E, Kressel H, Moss A, Montagne J. Computed tomography in the diagnosis of adrenal disease. American Journal of Roentgenology 1979;132(2):231–238.
  2. Vassiliadi D a, Tsagarakis S. Endocrine incidentalomas--challenges imposed by incidentally discovered lesions. Nat Rev Endocrinol 2011;7(11):668–80.
  3. Cawood TJ, Hunt PJ, O&apos;Shea D, Cole D, Soule S. Recommended evaluation of adrenal incidentalomas is costly, has high false-positive rates and confers a risk of fatal cancer that is similar to the risk of the adrenal lesion becoming malignant; time for a rethink? Eur J Endocrinol 2009;161(4):513–527.
  4. Grumbach MM, Biller BMK, Braunstein GD, Campbell KK, Aidan Carney J, Godley PA, Harris EL, Lee JKT, Oertel YC, Posner MC, Schlechte JA, Wieand S, Marciel K, Carney JA, Godley PA, Harris EL, Lee JKT, Oertel YC, Posner MC, Schlechte JA, Wieand HS. Management of the clinically inapparent adrenal mass (“incidentaloma”). In: Annals of Internal Medicine.Vol 138.; 2003:424–429.
  5. Terzolo M, Stigliano A, Chiodini I, Loli P, Furlani L, Arnaldi G, Reimondo G, Pia A, Toscano V, Zini M, Borretta G, Papini E, Garofalo P, Allolio B, Dupas B, Mantero F, Tabarin A. AME position statement on adrenal incidentaloma. Eur J Endocrinol 2011;164(6):851–870.
  6. Young WF. The Incidentally Discovered Adrenal Mass. New England Journal of Medicine 2007;356(6):601–610.
  7. Nawar R. Adrenal incidentalomas -- a continuing management dilemma. Endocrine Related Cancer 2005;12(3):585–598.
  8. Barzon L, Sonino N, Fallo F, Palù G, Boscaro M. Prevalence and natural history of adrenal incidentalomas. Eur J Endocrinol 2003;149(4):273–285.
  9. Ebbehoj A, Li D, Kaur RJ, Zhang C, Singh S, Li T, Atkinson E, Achenbach S, Khosla S, Arlt W, Young WF, Rocca WA, Bancos I. Epidemiology of adrenal tumours in Olmsted County, Minnesota, USA: a population-based cohort study. Lancet Diabetes Endocrinol 2020;8(11):894–902.
  10. van den Broek J, Geenen R, Heijnen L, Kobus C, Schreurs H. Adrenal Incidentalomas During Diagnostic Work-up of Colorectal Cancer Patients: What is the Risk of Metastases? Ann Surg Oncol 2018;25(7):1986–1991.
  11. Rineheart JF WO and CW. Adenomatous hyperplasia of the adrenal cortex associated with essential hypertension. Arch Pathol (Chic) 1941;(34):1031–1034.
  12. RUSSI S, BLUMENTHAL HT, GRAY SH. Small adenomas of the adrenal cortex in hypertension and diabetes. Arch Intern Med (Chic) 1945;76:284–91.
  13. COMMONS RR, CALLAWAY CP. Adenomas of the adrenal cortex. Arch Intern Med (Chic) 1948;81(1):37–41.
  14. Schroeder H. Clinical types - the endocrine hypertensive syndrome. In: Schroeder H, ed. Hypertensive Diseases: Causes and Control. Philadelphia: Lea & Febiger; 1953:295–333.
  15. Dévényi I. Possibility of normokalaemic primary aldosteronism as reflected in the frequency of adrenal cortical adenomas. J Clin Pathol 1967;20(1):49 LP – 51.
  16. Salomon MI, Tchertkoff V. Adrenal adenoma and hypertension. Ann Intern Med 1972;76(4):668–669.
  17. Hedeland H, Östberg G, Hökfelt B. ON THE PREVALENCE OF ADRENOCORTICAL ADENOMAS IN AN AUTOPSY MATERIAL IN RELATION TO HYPERTENSION AND DIABETES. Acta Med Scand 1968;184(1‐6):211–214.
  18. Yamada EY, Fukunaga FH. Adrenal Adenoma and Hypertension a Study in the Japanese in Hawaii. Jpn Heart J 1969;10(1):11–19.
  19. Granger P, Genest J. Autopsy study of adrenals in unselected normotensive and hypertensive patients. Can Med Assoc J 1970;103(1):34–36.
  20. Russell RP, Masi AT, Richter ED. Adrenal cortical adenomas and hypertension: A clinical pathologic analysis of 690 cases with matched contbols and a review of the literature. Medicine (United States) 1972;51(3):211–225.
  21. Abecassis M, McLoughlin MJ, Langer B, Kudlow JE. Serendipitous adrenal masses: Prevalence, significance, and management. The American Journal of Surgery 1985;149(6):783–788.
  22. Meagher AP, Hugh TB, Casey JH, Chisholm DJ, Farrell JC, Yeates M. PRIMARY ADRENAL TUMOURS – A TEN‐YEAR EXPERIENCE. Australian and New Zealand Journal of Surgery 1988;58(6):457–462.
  23. Reinhard C, Saeger W, Schubert B. Adrenocortical nodules in post-mortem series. Development, functional significance, and differentiation from adenomas. Gen Diagn Pathol 1995;141(3–4):203–208.
  24. Kloos RT, Gross MD, Francis IR, Korobkin M, Shapiro B. Incidentally discovered adrenal masses. Endocr Rev 1995;16(4):460–484.
  25. Masumori N, Adachi H, Noda Y, Tsukamoto T. Detection of adrenal and retroperitoneal masses in a general health examination system. Urology 1998;52(4):572–576.
  26. Glazer H, Weyman P, Sagel S, Levitt R, McClennan B. Nonfunctioning adrenal masses: incidental discovery on computed tomography. American Journal of Roentgenology 1982;139(1):81–85.
  27. Prinz RA, Brooks MH, Churchill R, Graner JL, Lawrence AM, Paloyan E, Sparagana M. Incidental Asymptomatic Adrenal Masses Detected by Computed Tomographic Scanning: Is Operation Required? JAMA: The Journal of the American Medical Association 1982;248(6):701–704.
  28. Belldegrun A, Hussain S, Seltzer SE, Loughlin KR, Gittes RF, Richie JP. Incidentally discovered mass of the adrenal gland. Surg Gynecol Obstet 1986;163(3):203–208.
  29. Herrera MF, Grant CS, van Heerden JA, Sheedy PF, Ilstrup DM. Incidentally discovered adrenal tumors: an institutional perspective. Surgery 1991;110(6):1014–21.
  30. Caplan RH, Strutt PJ, Wickus GG. Subclinical Hormone Secretion by Incidentally Discovered Adrenal Masses. Archives of Surgery 1994;129(3):291–296.
  31. Bovio S, Cataldi A, Reimondo G, Sperone P, Novello S, Berruti A, Borasio P, Fava C, Dogliotti L, Scagliotti G V., Angeli A, Terzolo M. Prevalence of adrenal incidentaloma in a contemporary computerized tomography series. J Endocrinol Invest 2006;29(4):298–302.
  32. Song JH, Chaudhry FS, Mayo-Smith WW. The incidental adrenal mass on CT: Prevalence of adrenal disease in 1,049 consecutive adrenal masses in patients with no known malignancy. American Journal of Roentgenology 2008;190(5):1163–1168.
  33. Mantero F, Terzolo M, Arnaldi G, Osella G, Masini AM, Ali A, Giovagnetti M, Opocher G, Angeli A. A survey on adrenal incidentaloma in Italy. Study Group on Adrenal Tumors of the Italian Society of Endocrinology. J Clin Endocrinol Metab 2000;85(2):637–644.
  34. Jing Y, Hu J, Luo R, Mao Y, Luo Z, Zhang M, Yang J, Song Y, Feng Z, Wang Z, Cheng Q, Ma L, Yang Y, Zhong L, Du Z, Wang Y, Luo T, He W, Sun Y, Lv F, Li Q, Yang S. Prevalence and Characteristics of Adrenal Tumors in an Unselected Screening Population. https://doi.org/10.7326/M22-1619 2022;175(10):1383–1391.
  35. Mayer SK, Oligny LL, Deal C, Yazbeck S, Gagné IN, Blanchard H. Childhood adrenocortical tumors: Case series and reevaluation of prognosis - A 24-year experience. J Pediatr Surg 1997;32(6):911–915.
  36. Angeli A, Osella G, Ali A, Terzolo M. Adrenal incidentaloma: An overview of clinical and epidemiological data from the national italian study group. Horm Res Paediatr 1997;47(4–6):279–283.
  37. Aron D, Terzolo M, Cawood TJ. Adrenal incidentalomas. Best Pract Res Clin Endocrinol Metab 2012;26(1):69–82.
  38. Hsiao HP, Kirschner LS, Bourdeau I, Keil MF, Boikos SA, Verma S, Robinson-White AJ, Nesterova M, Lacroix A, Stratakis CA. Clinical and genetic heterogeneity, overlap with other tumor syndromes, and atypical glucocorticoid hormone secretion in adrenocorticotropin-independent macronodular adrenal hyperplasia compared with other adrenocortical tumors. J Clin Endocrinol Metab 2009;94(8):2930–2937.
  39. Reznik Y, Lefebvre H, Rohmer V, Charbonnel B, Tabarin A, Rodien P, Lecomte P, Bardet S, Coffin C, Mahoudeau J. Aberrant adrenal sensitivity to multiple ligands in unilateral incidentaloma with subclinical autonomous cortisol hypersecretion: a prospective clinical study. Clin Endocrinol (Oxf) 2004;61(3):311–319.
  40. Lenders JWM, Duh Q-Y, Eisenhofer G, Gimenez-Roqueplo A-P, Grebe SKG, Murad MH, Naruse M, Pacak K, Young WF. Pheochromocytoma and Paraganglioma: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2014;99(6):1915–1942.
  41. Kopetschke R, Slisko M, Kilisli A, Tuschy U, Wallaschofski H, Fassnacht M, Ventz M, Beuschlein F, Reincke M, Reisch N, Quinkler M. Frequent incidental discovery of phaeochromocytoma: Data from a German cohort of 201 phaeochromocytoma. Eur J Endocrinol 2009;161(2):355–361.
  42. Funder JW, Carey RM, Mantero F, Murad MH, Reincke M, Shibata H, Stowasser M, Young WF. The Management of Primary Aldosteronism: Case Detection, Diagnosis, and Treatment: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2016;101(5):1889–1916.
  43. Piaditis G, Markou A, Papanastasiou L, Androulakis II, Kaltsas G. Progress in aldosteronism: A review of the prevalence of primary aldosteronism in pre-hypertension and hypertension. Eur J Endocrinol 2015;172(5):R191–R203.
  44. Médeau V, Moreau F, Trinquart L, Clemessy M, Wémeau JL, Vantyghem MC, Plouin PF, Reznik Y. Clinical and biochemical characteristics of normotensive patients with primary aldosteronism: A comparison with hypertensive cases. Clin Endocrinol (Oxf) 2008;69(1):20–28.
  45. Piaditis GP, Kaltsas GA, Androulakis II, Gouli A, Makras P, Papadogias D, Dimitriou K, Ragkou D, Markou A, Vamvakidis K, Zografos G, Chrousos G. High prevalence of autonomous cortisol and aldosterone secretion from adrenal adenomas. Clin Endocrinol (Oxf) 2009;71(6):772–778.
  46. Sherlock M, Scarsbrook A, Abbas A, Fraser S, Limumpornpetch P, Dineen R, Stewart PM. Adrenal Incidentaloma. Endocr Rev 2020;41(6). doi:10.1210/ENDREV/BNAA008.
  47. Liao Z, Gao Y, Zhao Y, Wang Z, Wang X, Zhou J, Zhang Y. Pure androgen-secreting adrenal tumor (PASAT): A rare case report of bilateral PASATs and a systematic review. Front Endocrinol (Lausanne) 2023;14:678.
  48. Mansmann G, Lau J, Balk E, Rothberg M, Miyachi Y, Bornstein SR. The clinically inapparent adrenal mass: Update in diagnosis and management. Endocr Rev 2004;25(2):309–340.
  49. Zeiger MA, Siegelman SS, Hamrahian AH. Medical and surgical evaluation and treatment of adrenal incidentalomas. Journal of Clinical Endocrinology and Metabolism 2011;96(7):2004–2015.
  50. Fallo F, Barzon L, Boscaro M, Sonino N. Coexistence of aldosteronoma and contralateral nonfunctioning adrenal adenoma in primary aldosteronism. Am J Hypertens 1997;10(4 I):476–478.
  51. Satoh F, Murakami O, Takahashi K, Ueno J, Nishikawa T, Abe K, Mouri T, Sasano H. Double adenomas with different pathological and hormonal features in the left adrenal gland of a patient with Cushing’s syndrome. Clin Endocrinol (Oxf) 1997;46(2):227–234.
  52. Morimoto S, Sasaki S, Moriguchi J, Miki S, Kawa T, Nakamura K, Fujita H, Itoh H, Nakata T, Takeda K, Nakagawa M. Unique association of pheochromocytoma with contralateral nonfunctioning adrenal cortical adenoma. Am J Hypertens 1998;11(1):117–121.
  53. Chortis V, May CJH, Skordilis K, Ayuk J, Arlt W, Crowley RK. Double trouble: Two cases of dual adrenal pathologies in one adrenal mass. Endocrinol Diabetes Metab Case Rep 2019;2019(1). doi:10.1530/EDM-18-0151.
  54. Dobbie JW. Adrenocortical nodular hyperplasia: The ageing adrenal. J Pathol 1969;99(1):1–18.
  55. Beuschlein F, Reincke M, Allolio B, Karl M, Travis WD, Jaursch-Hancke C, Abdelhamid S, Chrousos GP. Clonal Composition of Human Adrenocortical Neoplasms. Cancer Res 1994;54(18):4927–4932.
  56. Gicquel C, Leblond-Francillard M, Bertagna X, Louvel A, Chapuis Y, Luton JP, Girard F, Le Bouc Y. Clonal analysis of human adrenocortical carcinomas and secreting adenomas. Clin Endocrinol (Oxf) 1994;40(4):465–477.
  57. Pillion DJ, Arnold P, Yang M, Stockard CR, Grizzle WE. Receptors for insulin and insulin-like growth factor-I in the human adrenal gland. Biochem Biophys Res Commun 1989;165(1):204–211.
  58. Reincke M, Faßnacht M, Väth S, Mora P, Allolio B. Adrenal incidentalomas: A manifestation of the metabolic syndrome? In: Endocrine Research.Vol 22.; 1996:757–761.
  59. Angeli A, Terzolo M. Editorial: Adrenal incidentaloma - A modern disease with old complications. Journal of Clinical Endocrinology and Metabolism 2002;87(11):4869–4871.
  60. Vassiliadi DA, Tzanela M, Tsatlidis V, Margelou E, Tampourlou M, Mazarakis N, Piaditis G, Tsagarakis S. Abnormal responsiveness to dexamethasone-suppressed CRH test in patients with bilateral adrenal incidentalomas. Journal of Clinical Endocrinology and Metabolism 2015;100(9):3478–3485.
  61. Bertagna X. Genetics of adrenal diseases in 2014: Genetics improves understanding of adrenocortical tumours. Nat Rev Endocrinol 2014;11(2):77–78.
  62. Lerario AM, Moraitis A, Hammer GD. Genetics and epigenetics of adrenocortical tumors. Mol Cell Endocrinol 2014;386(1–2):67–84.
  63. Bonnet-Serrano F, Bertherat J. Genetics of tumors of the adrenal cortex. Endocr Relat Cancer 2018;25(3):R131–R152.
  64. Zeiger M, Thompson G, Duh Q-Y, Hamrahian A, Angelos P, Elaraj D, Fishman E, Kharlip J. American Association of Clinical Endocrinologists and American Association of Endocrine Surgeons Medical Guidelines for the Management of Adrenal Incidentalomas. Endocrine Practice 2009;15(Supplement 1):1–20.
  65. Lau J, Balk E, Rothberg M, Ioannidis JP, DeVine D, Chew P, Kupelnick B, Miller K. Management of clinically inapparent adrenal mass. Evid Rep Technol Assess (Summ) 2002;(56):1–5.
  66. Fassnacht M, Tsagarakis S, Terzolo M, Tabarin A, Sahdev A, Newell-Price J, Pelsma I, Marina L, Lorenz K, Bancos I, Arlt W, Dekkers OM. European Society of Endocrinology clinical practice guidelines on the management of adrenal incidentalomas, in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol 2023;189(1). doi:10.1093/EJENDO/LVAD066.
  67. Pantalone KM, Gopan T, Remer EM, Faiman C, Ioachimescu AG, Levin HS, Siperstein A, Berber E, Shepardson LB, Bravo EL, Hamrahian AH. Change in adrenal mass size as a predictor of a malignant tumor. Endocr Pract 2010;16(4):577–587.
  68. Kaltsas G, Chrisoulidou A, Piaditis G, Kassi E, Chrousos G. Current status and controversies in adrenal incidentalomas. Trends in Endocrinology and Metabolism 2012;23(12):602–609.
  69. Blake MA, Holalkere NS, Boland GW. Imaging Techniques for Adrenal Lesion Characterization. Radiol Clin North Am 2008;46(1):65–78.
  70. Dinnes J, Bancos I, di Ruffano LF, Chortis V, Davenport C, Bayliss S, Sahdev A, Guest P, Fassnacht M, Deeks JJ, Arlt W. MANAGEMENT OF ENDOCRINE DISEASE: Imaging for the diagnosis of malignancy in incidentally discovered adrenal masses: a systematic review and meta-analysis. Eur J Endocrinol 2016;175(2):R51–R64.
  71. Hamrahian AH, Ioachimescu AG, Remer EM, Motta-Ramirez G, Bogabathina H, Levin HS, Reddy S, Gill IS, Siperstein A, Bravo EL. Clinical utility of noncontrast computed tomography attenuation value (hounsfield units) to differentiate adrenal adenomas/hyperplasias from nonadenomas: Cleveland clinic experience. Journal of Clinical Endocrinology and Metabolism 2005;90(2):871–877.
  72. Bancos I, Taylor AE, Chortis V, Sitch AJ, Jenkinson C, Davidge-Pitts CJ, Lang K, Tsagarakis S, Macech M, Riester A, Deutschbein T, Pupovac ID, Kienitz T, Prete A, Papathomas TG, Gilligan LC, Bancos C, Reimondo G, Haissaguerre M, Marina L, Grytaas MA, Sajwani A, Langton K, Ivison HE, Shackleton CHL, Erickson D, Asia M, Palimeri S, Kondracka A, Spyroglou A, Ronchi CL, Simunov B, Delivanis DA, Sutcliffe RP, Tsirou I, Bednarczuk T, Reincke M, Burger-Stritt S, Feelders RA, Canu L, Haak HR, Eisenhofer G, Dennedy MC, Ueland GA, Ivovic M, Tabarin A, Terzolo M, Quinkler M, Kastelan D, Fassnacht M, Beuschlein F, Ambroziak U, Vassiliadi DA, O’Reilly MW, Young WF, Biehl M, Deeks JJ, Arlt W, Glöckner S, Sinnott RO, Stell A, Fragoso MC, Pupovac ID, Cazenave S, Bertherat J, Libé R, Brugger C, Hahner S, Kroiss M, Ronchi CL, Vassiliadi DA, Basile V, Ingargiola E, Mannelli M, Ettaieb H, Haak HR, Kerkhofs TM, Feelders RA, Hofland J, Hofland LJ, Grytaas MA, Husebye ES, Ueland GA, Zawierucha M, Paiva I, Dennedy MC, Sherlock M, Crowley RK, Jonathan R, Sitch AJ, Giligan LC, Hughes BA, Ivison HE, Manolopoulos K, O’Neil DM, O’Reilly MW, Papathomas TG, Shackleton CHL, Taylor AE, Sutcliffe RP, Guest P, Skordilis K, Chang A, Davidge-Pitts CJ, Delivanis DA, Natt N, Nippoldt TB, Thomas M, Young WF. Urine steroid metabolomics for the differential diagnosis of adrenal incidentalomas in the EURINE-ACT study: a prospective test validation study. Lancet Diabetes Endocrinol 2020;8(9):773–781.
  73. Vilar L, da Conceição Freitas M, Canadas V, Albuquerque JL, Botelho CA, Egito CS, Arruda MJ, Moura e Silva L, Coelho CE, Casulari LA, Naves LA. Adrenal Incidentalomas: Diagnostic Evaluation and Long-Term Follow-Up. Endocrine Practice 2008;14(3):269–278.
  74. Marty M, Gaye D, Perez P, Auder C, Nunes ML, Ferriere A, Haissaguerre M, Tabarin A. Diagnostic accuracy of computed tomography to identify adenomas among adrenal incidentalomas in an endocrinological population. Eur J Endocrinol 2018;178(5):439–446.
  75. Schloetelburg W, Ebert I, Petritsch B, Weng AM, Dischinger U, Kircher S, Buck AK, Bley TA, Deutschbein T, Fassnacht M. Adrenal wash-out CT: moderate diagnostic value in distinguishing benign from malignant adrenal masses. Eur J Endocrinol 2022;186(2):183–193.
  76. Hong AR, Kim JH, Park KS, Kim KY, Lee JH, Kong SH, Lee SY, Shin CS, Kim SW, Kim SY. Optimal follow-up strategies for adrenal incidentalomas: reappraisal of the 2016 ESE-ENSAT guidelines in real clinical practice. Eur J Endocrinol 2017;177(6):475–483.
  77. Korobkin M, Brodeur FJ, Francis IR, Quint LE, Dunnick NR, Londy F. CT time-attenuation washout curves of adrenal adenomas and nonadenomas. American Journal of Roentgenology 1998;170(3):747–752.
  78. Caoili EM, Korobkin M, Francis IR, Cohan RH, Platt JF, Dunnick NR, Raghupathi KI. Adrenal masses: Characterization with combined unenhanced delayed enhanced CT. Radiology 2002;222(3):629–633.
  79. Motta-Ramirez GA, Remer EM, Herts BR, Gill IS, Hamrahian AH. Comparison of CT findings in symptomatic and incidentally discovered pheochromocytomas. American Journal of Roentgenology 2005;185(3):684–688.
  80. Corwin MT, Badawy M, Caoili EM, Carney BW, Colak C, Elsayes KM, Gerson R, Klimkowski SP, McPhedran R, Pandya A, Pouw ME, Schieda N, Song JH, Remer EM. Incidental Adrenal Nodules in Patients Without Known Malignancy: Prevalence of Malignancy and Utility of Washout CT for Characterization-A Multiinstitutional Study. American Journal of Roentgenology 2022;219(5):804–813.
  81. Chambre C, McMurray E, Baudry C, Lataud M, Guignat L, Gaujoux S, Lahlou N, Guibourdenche J, Tissier F, Sibony M, Dousset B, Bertagna X, Bertherat J, Legmann P, Groussin L. The 10 Hounsfield units unenhanced computed tomography attenuation threshold does not apply to cortisol secreting adrenocortical adenomas.; 2015:325–332.
  82. Blake MA, Kalra MK, Sweeney AT, Lucey BC, Maher MM, Sahani D V, Halpern EF, Mueller PR, Hahn PF, Boland GW. Distinguishing benign from malignant adrenal masses: Multi-detector row CT protocol with 10-minute delay. Radiology 2006;238(2):578–585.
  83. Wickramarachchi BN, Meyer-Rochow GY, McAnulty K, Conaglen J V., Elston MS. Adherence to adrenal incidentaloma guidelines is influenced by radiology report recommendations. ANZ J Surg 2016;86(6):483–486.
  84. de Haan RR, Schreuder MJ, Pons E, Visser JJ. Adrenal Incidentaloma and Adherence to International Guidelines for Workup Based on a Retrospective Review of the Type of Language Used in the Radiology Report. Journal of the American College of Radiology 2019;16(1):50–55.
  85. Watari J, Vekaria S, Lin Y, Patel M, Kim H, Kang F, Lubitz S, Beninato T, Laird AM. Radiology report language positively influences adrenal incidentaloma guideline adherence. Am J Surg 2022;223(2):231–236.
  86. Korobkin M, Francis IR, Kloos RT, Dunnick NR. The incidental adrenal mass. Radiol Clin North Am 1996;34(5):1037–54.
  87. Boland GWL. Adrenal imaging: Why, when, what, and how? Part 3. The algorithmic approach to definitive characterization of the adrenal incidentaloma. American Journal of Roentgenology 2011;196(2):W109–W111.
  88. McDermott S, O’Connor OJ, Cronin CG, Blake MA. Radiological evaluation of adrenal incidentalomas - Current methods and future prospects. Best Pract Res Clin Endocrinol Metab 2012;26(1):21–33.
  89. Elsayes KM, Menias CO, Siegel CL, Narra VR, Kanaan Y, Hussain HK. Magnetic resonance characterization of pheochromocytomas in the abdomen and pelvis: Imaging findings in 18 surgically proven cases. J Comput Assist Tomogr 2010;34(4):548–553.
  90. Nunes ML, Rault A, Teynie J, Valli N, Guyot M, Gaye D, Belleannee G, Tabarin A. 18F-FDG PET for the identification of adrenocortical carcinomas among indeterminate adrenal tumors at computed tomography scanning. World J Surg 2010;34(7):1506–1510.
  91. Tessonnier L, Sebag F, Palazzo FF, Colavolpe C, De Micco C, Mancini J, Conte-Devolx B, Henry JF, Mundler O, Taïeb D. Does 18F-FDG PET/CT add diagnostic accuracy in incidentally identified non-secreting adrenal tumours?; 2008:2018–2025.
  92. Boland GWL, Blake MA, Holalkere NS, Hahn PF. PET/CT for the characterization of adrenal masses in patients with cancer: Qualitative versus quantitative accuracy in 150 consecutive patients. American Journal of Roentgenology 2009;192(4):956–962.
  93. Groussin L, Bonardel G, Silvéra S, Tissier F, Coste J, Abiven G, Libé R, Bienvenu M, Alberini JL, Salenave S, Bouchard P, Bertherat J, Dousset B, Legmann P, Richard B, Foehrenbach H, Bertagna X, Tenenbaum F. 18F-Fluorodeoxyglucose positron emission tomography for the diagnosis of adrenocortical tumors: A prospective study in 77 operated patients. Journal of Clinical Endocrinology and Metabolism 2009;94(5):1713–1722.
  94. Sharma P, Singh H, Dhull VVS, Suman KC S, Kumar A, Bal C, Kumar R. Adrenal Masses of Varied Etiology: Anatomical and Molecular Imaging Features on PET-CT. Clinical nuclear … 2014;00(00):1–10.
  95. Havekes B, King K, Lai EW, Romijn JA, Corssmit EPM, Pacak K. New imaging approaches to phaeochromocytomas and paragangliomas. Clin Endocrinol (Oxf) 2010;72(2):137–145.
  96. Gross MD, Shapiro B, Francis IR, Glazer GM, Bree RL, Arcomano MA, Schteingart DE, McLeod MK, Sanfield JA, Thompson NW, Falke THM, Sandler MP. Scintigraphic evaluation of clinically silent adrenal masses. Journal of Nuclear Medicine 1994;35(7):1145–1154.
  97. Hahner S, Stuermer A, Kreissl M, Reiners C, Fassnacht M, Haenscheid H, Beuschlein F, Zink M, Lang K, Allolio B, Schirbel A. [123I]Iodometomidate for molecular imaging of adrenocortical cytochrome P450 family 11B enzymes. Journal of Clinical Endocrinology and Metabolism 2008;93(6):2358–2365.
  98. Nieman LK, Biller BMK, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM, Edwards H. The diagnosis of Cushing’s syndrome: An endocrine society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism 2008;93(5):1526–1540.
  99. Morelli V, Eller-Vainicher C, Salcuni AS, Coletti F, Iorio L, Muscogiuri G, Della Casa S, Arosio M, Ambrosi B, Beck-Peccoz P, Chiodini I. Risk of new vertebral fractures in patients with adrenal incidentaloma with and without subclinical hypercortisolism: A multicenter longitudinal study. John Wiley & Sons, Ltd; 2011:1816–1821.
  100. Olsen H, Kjellbom A, Löndahl M, Lindgren O. Suppressed ACTH Is Frequently Unrelated to Autonomous Cortisol Secretion in Patients With Adrenal Incidentalomas. J Clin Endocrinol Metab 2019;104(2):506–512.
  101. Fassnacht M, Arlt W, Bancos I, Dralle H, Newell-Price J, Sahdev A, Tabarin A, Terzolo M, Tsagarakis S, Dekkers OM. Management of adrenal incidentalomas: European Society of Endocrinology Clinical Practice Guideline in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol 2016;175(2):G1–G34.
  102. Tabarin A, Bardet S, Bertherat J, Dupas B, Chabre O, Hamoir E, Laurent F, Tenenbaum F, Cazalda M, Lefebvre H, Valli N, Rohmer V. Exploration and management of adrenal incidentalomas. French Society of Endocrinology Consensus. Ann Endocrinol (Paris) 2008;69(6):487–500.
  103. Morelli V, Masserini B, Salcuni AS, Eller-Vainicher C, Savoca C, Viti R, Coletti F, Guglielmi G, Battista C, Iorio L, Beck-Peccoz P, Ambrosi B, Arosio M, Scillitani A, Chiodini I. Subclinical hypercortisolism: Correlation between biochemical diagnostic criteria and clinical aspects. Clin Endocrinol (Oxf) 2010;73(2):161–166.
  104. Barzon L, Scaroni C, Sonino N, Fallo F, Paoletta A, Boscaro M. Risk Factors and Long-Term Follow-Up of Adrenal Incidentalomas1. J Clin Endocrinol Metab 1999;84(2):520–526.
  105. Eller-Vainicher C, Morelli V, Salcuni AS, Torlontano M, Coletti F, Iorio L, Cuttitta A, Ambrosio A, Vicentini L, Carnevale V, Beck-Peccoz P, Arosio M, Ambrosi B, Scillitani A, Chiodini I. Post-surgical hypocortisolism after removal of an adrenal incidentaloma: Is it predictable by an accurate endocrinological work-up before surgery? Eur J Endocrinol 2010;162(1):91–99.
  106. Eller-Vainicher C, Morelli V, Salcuni AS, Battista C, Torlontano M, Coletti F, Iorio L, Cairoli E, Beck-Peccoz P, Arosio M, Ambrosi B, Scillitani A, Chiodini I. Accuracy of several parameters of hypothalamic-pituitary-adrenal axis activity in predicting before surgery the metabolic effects of the removal of an adrenal incidentaloma. Eur J Endocrinol 2010;163(6):925–935.
  107. Chiodini I, Morelli V, Salcuni AS, Eller-Vainicher C, Torlontano M, Coletti F, Iorio L, Cuttitta A, Ambrosio A, Vicentini L, Pellegrini F, Copetti M, Beck-Peccoz P, Arosio M, Ambrosi B, Trischitta V, Scillitani A. Beneficial metabolic effects of prompt surgical treatment in patients with an adrenal incidentaloma causing biochemical hypercortisolism. Journal of Clinical Endocrinology and Metabolism 2010;95(6):2736–2745.
  108. Di Dalmazi G, Vicennati V, Garelli S, Casadio E, Rinaldi E, Giampalma E, Mosconi C, Golfieri R, Paccapelo A, Pagotto U, Pasquali R. Cardiovascular events and mortality in patients with adrenal incidentalomas that are either non-secreting or associated with intermediate phenotype or subclinical Cushing’s syndrome: A 15-year retrospective study. Lancet Diabetes Endocrinol 2014;2(5):396–405.
  109. Debono M, Bradburn M, Bull M, Harrison B, Ross RJ, Newell-Price J. Cortisol as a marker for increased mortality in patients with incidental adrenocortical adenomas. Journal of Clinical Endocrinology and Metabolism 2014;99(12):4462–4470.
  110. Tsagarakis S, Vassiliadi D, Thalassinos N. Endogenous subclinical hypercortisolism: Diagnostic uncertainties and clinical implications. J Endocrinol Invest 2006;29(5):471–482.
  111. Theodoraki A, Khoo B, Hamda A, Schwappach A, Perera S, Vanderpump MPJ, Bouloux PM. Outcomes in 125 individuals with adrenal incidentalomas from a single centre. A retrospective assessment of the 1mg overnight and low dose dexamethasone suppression tests. Hormone and Metabolic Research 2011;43(13):962–969.
  112. Tsagarakis S, Roboti C, Kokkoris P, Vasiliou V, Alevizaki C, Thalassinos N. Elevated post-dexamethasone suppression cortisol concentrations correlate with hormonal alterations of the hypothalamo-pituitary adrenal axis in patients with adrenal incidentalomas. Clin Endocrinol (Oxf) 1998;49(2):165–171.
  113. Lopez AG, Fraissinet F, Lefebvre H, Brunel V, Ziegler F. Pharmacological and analytical interference in hormone assays for diagnosis of adrenal incidentaloma. Ann Endocrinol (Paris) 2019;80(4):250–258.
  114. Liu MS, Lou Y, Chen H, Wang YJ, Zhang ZW, Li P, Zhu DL. Performance of DHEAS as a Screening Test for Autonomous Cortisol Secretion in Adrenal Incidentalomas: A Prospective Study. J Clin Endocrinol Metab 2022;107(5):e1789–e1796.
  115. Dennedy MC, Annamalai AK, Prankerd-Smith O, Freeman N, Vengopal K, Graggaber J, Koulouri O, Powlson AS, Shaw A, Halsall DJ, Gurnell M. Low DHEAS: A Sensitive and Specific Test for the Detection of Subclinical Hypercortisolism in Adrenal Incidentalomas. J Clin Endocrinol Metab 2017;102(3):786–792.
  116. Carafone LE, Zhang CD, Li D, Lazik N, Hamidi O, Hurtado MD, Young WF, Thomas MA, Dy BM, Lyden ML, Foster TR, McKenzie TJ, Bancos I. Diagnostic Accuracy of Dehydroepiandrosterone Sulfate and Corticotropin in Autonomous Cortisol Secretion. Biomedicines 2021;9(7). doi:10.3390/BIOMEDICINES9070741.
  117. Hána V, Ježková J, Kosák M, Kršek M, Hána V, Hill M. Novel GC-MS/MS Technique Reveals a Complex Steroid Fingerprint of Subclinical Hypercortisolism in Adrenal Incidentalomas. J Clin Endocrinol Metab 2019;104(8):3545–3556.
  118. Araujo-Castro M, Casals G, Hanzu FA, Pascual-Corrales E, García Cano AM, Lanza VF, Luis del Rey Mejías Á, Marchan M, Escobar-Morreale HF, Valderrabano P. Characterisation of the urinary steroid profile of patients with nonfunctioning adrenal incidentalomas: A matched controlled cross-sectional study. Clin Endocrinol (Oxf) 2023;98(2):165–176.
  119. Berke K, Constantinescu G, Masjkur J, Kimpel O, Dischinger U, Peitzsch M, Kwapiszewska A, Dobrowolski P, Nölting S, Reincke M, Beuschlein F, Bornstein SR, Prejbisz A, Lenders JWM, Fassnacht M, Eisenhofer G. Plasma Steroid Profiling in Patients with Adrenal Incidentaloma. Journal of Clinical Endocrinology and Metabolism 2022;107(3):E1181–E1192.
  120. Grozinsky-Glasberg S, Szalat A, Benbassat CA, Gorshtein A, Weinstein R, Hirsch D, Shraga-Slutzky I, Tsvetov G, Gross DJ, Shimon I. Clinically silent chromaffin-cell tumors: Tumor characteristics and long-term prognosis in patients with incidentally discovered pheochromocytomas. J Endocrinol Invest 2010;33(10):739–744.
  121. Lenders JWM, Pacak K, Walther MM, Marston Linehan W, Mannelli M, Friberg P, Keiser HR, Goldstein DS, Eisenhofer G. Biochemical diagnosis of pheochromocytoma: Which test is best? J Am Med Assoc 2002;287(11):1427–1434.
  122. Eisenhofer G, Prejbisz A, Peitzsch M, Pamporaki C, Masjkur J, Rogowski-Lehmann N, Langton K, Tsourdi E, Peczkowska M, Fliedner S, Deutschbein T, Megerle F, Timmers HJLM, Sinnott R, Beuschlein F, Fassnacht M, Januszewicz A, Lenders JWM. Biochemical Diagnosis of Chromaffin Cell Tumors in Patients at High and Low Risk of Disease: Plasma versus Urinary Free or Deconjugated O-Methylated Catecholamine Metabolites. Clin Chem 2018;64(11):1646–1656.
  123. Sane T, Schalin-Jäntti C, Raade M. Is biochemical screening for pheochromocytoma in adrenal incidentalomas expressing low unenhanced attenuation on computed tomography necessary? Journal of Clinical Endocrinology and Metabolism 2012;97(6):2077–2083.
  124. Canu L, Van Hemert JAW, Kerstens MN, Hartman RP, Khanna A, Kraljevic I, Kastelan D, Badiu C, Ambroziak U, Tabarin A, Haissaguerre M, Buitenwerf E, Visser A, Mannelli M, Arlt W, Chortis V, Bourdeau I, Gagnon N, Buchy M, Borson-Chazot F, Deutschbein T, Fassnacht M, Hubalewska-Dydejczyk A, Motyka M, Rzepka E, Casey RT, Challis BG, Quinkler M, Vroonen L, Spyroglou A, Beuschlein F, Lamas C, Young WF, Bancos I, Timmers HJLM. CT Characteristics of Pheochromocytoma: Relevance for the Evaluation of Adrenal Incidentaloma. Journal of Clinical Endocrinology and Metabolism 2018;104(2):312–318.
  125. Haissaguerre M, Courel M, Caron P, Denost S, Dubessy C, Gosse P, Appavoupoulle V, Belleannée G, Jullié ML, Montero-Hadjadje M, Yon L, Corcuff JB, Fagour C, Mazerolles C, Wagner T, Nunes ML, Anouar Y, Tabarin A. Normotensive incidentally discovered pheochromocytomas display specific biochemical, cellular, and molecular characteristics. Journal of Clinical Endocrinology and Metabolism 2013;98(11):4346–4354.
  126. Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano TJ, Hammer GD. Adrenocortical carcinoma. Endocr Rev 2014;35(2):282–326.
  127. Phornphutkul C, Okubo T, Wu K, Harel Z, Tracy TF, Pinar H, Chen S, Gruppuso PA, Goodwin G. Aromatase P450 expression in a feminizing adrenal adenoma presenting as isosexual precocious puberty. Journal of Clinical Endocrinology and Metabolism 2001;86(2):649–652.
  128. Goto T, Murakami O, Sato F, Haraguchi M, Yokoyama K, Sasano H. Oestrogen producing adrenocortical adenoma: Clinical, biochemical and immunohistochemical studies. Clin Endocrinol (Oxf) 1996;45(5):643–648.
  129. Fukushima A, Okada Y, Tanikawa T, Kawahara C, Misawa H, Kanda K, Morita E, Sasano H, Tanaka Y. Virilizing adrenocortical adenoma with Cushing’s syndrome, thyroid papillary carcinoma and hypergastrinemia in a middle-aged woman. Endocr J 2003;50(2):179–187.
  130. Rodríguez-Gutiérrez R, Bautista-Medina MA, Teniente-Sanchez AE, Zapata-Rivera MA, Montes-Villarreal J. Pure Androgen-Secreting Adrenal Adenoma Associated with Resistant Hypertension. Case Rep Endocrinol 2013;2013:1–4.
  131. Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet 2014;383(9935):2152–67.
  132. Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, Husebye ES, Merke DP, Murad MH, Stratakis CA, Torpy DJ. Diagnosis and treatment of primary adrenal insufficiency: An endocrine society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism 2016;101(2):364–389.
  133. Quayle FJ, Spitler JA, Pierce RA, Lairmore TC, Moley JF, Brunt LM. Needle biopsy of incidentally discovered adrenal masses is rarely informative and potentially hazardous. Surgery 2007;142(4):497–504.
  134. Bancos I, Tamhane S, Shah M, Delivanis DA, Alahdab F, Arlt W, Fassnacht M, Murad MH. Diagnosis of endocrine disease: The diagnostic performance of adrenal biopsy: A systematic review and meta-analysis. Eur J Endocrinol 2016;175(2):R65–R80.
  135. Mazzaglia PJ, Monchik JM. Limited value of adrenal biopsy in the evaluation of adrenal neoplasm: a decade of experience. Arch Surg 2009;144(5):465–470.
  136. Lumachi F, Borsato S, Tregnaghi A, Marino F, Fassina A, Zucchetta P, Marzola MC, Cecchin D, Bui F, Iacobone M, Favia G. High risk of malignancy in patients with incidentally discovered adrenal masses: Accuracy of adrenal imaging and image-guided fine-needle aspiration cytology. Tumori 2007;93(3):269–274.
  137. Harisinghani MG, Maher MM, Hahn PF, Gervais DA, Jhaveri K, Varghese J, Mueller PR. Predictive value of benign percutaneous adrenal biopsies in oncology patients. Clin Radiol 2002;57(10):898–901.
  138. Welch TJ, Sheedy PF, Stephens DH, Johnson CM, Swensen SJ. Percutaneous adrenal biopsy: Review of a 10-year experience. Radiology 1994;193(2):341–344.
  139. Vanderveen KA, Thompson SM, Callstrom MR, Young WF, Grant CS, Farley DR, Richards ML, Thompson GB. Biopsy of pheochromocytomas and paragangliomas: Potential for disaster. Surgery 2009;146(6):1158–1166.
  140. Fassnacht M, Allolio B. Clinical management of adrenocortical carcinoma. Best Pract Res Clin Endocrinol Metab 2009;23(2):273–289.
  141. Sutton St. JM, Sheps SG, Lie JT. Prevalence of clinically unsuspected pheochromocytoma. Review of a 50-year autopsy series. Mayo Clin Proc 1981;56(6):354–360.
  142. Bülow B, Jansson S, Juhlin C, Steen L, Thorén M, Wahrenberg H, Valdemarsson S, Wängberg B, Ahrén B. Adrenal incidentaloma - Follow up results from a Swedish prospective study. Eur J Endocrinol 2006;154(3):419–423.
  143. Bernini GP, Moretti A, Oriandini C, Bardini M, Taurino C, Salvetti A. Long-term morphological and hormonal follow-up in a single unit on 115 patients with adrenal incidentalomas. Br J Cancer 2005;92(6):1104–1109.
  144. Terzolo M, Bovio S, Reimondo G, Pia A, Osella G, Borretta G, Angeli A. Subclinical Cushing’s syndrome in adrenal incidentalomas. Endocrinol Metab Clin North Am 2005;34(2):423–439.
  145. Belmihoub I, Silvera S, Sibony M, Dousset B, Legmann P, Bertagna X, Bertherat J, Assié G. From benign adrenal incidentaloma to adrenocortical carcinoma: An exceptional random event. Eur J Endocrinol 2017;176(6):K15–K19.
  146. Rebielak ME, Wolf MR, Jordan R, Oxenberg JC. Adrenocortical carcinoma arising from an adrenal adenoma in a young adult female. J Surg Case Rep 2019;2019(7):rjz200.
  147. Ronchi CL, Sbiera S, Leich E, Henzel K, Rosenwald A, Allolio B, Fassnacht M. Single Nucleotide Polymorphism Array Profiling of Adrenocortical Tumors - Evidence for an Adenoma Carcinoma Sequence? Veitia RA, ed. PLoS One 2013;8(9):e73959.
  148. Assié G, Letouzé E, Fassnacht M, Jouinot A, Luscap W, Barreau O, Omeiri H, Rodriguez S, Perlemoine K, René-Corail F, Elarouci N, Sbiera S, Kroiss M, Allolio B, Waldmann J, Quinkler M, Mannelli M, Mantero F, Papathomas T, De Krijger R, Tabarin A, Kerlan V, Baudin E, Tissier F, Dousset B, Groussin L, Amar L, Clauser E, Bertagna X, Ragazzon B, Beuschlein F, Libé R, de Reyniès A, Bertherat J. Integrated genomic characterization of adrenocortical carcinoma. Nat Genet 2014;46(6):607–612.
  149. Libè R, Dall’Asta C, Barbetta L, Baccarelli A, Beck-Peccoz P, Ambrosi B. Long-term follow-up study of patients with adrenal incidentalomas. Eur J Endocrinol 2002;147(4):489–494.
  150. Androulakis II, Kaltsas G, Piaditis G, Grossman AB. The clinical significance of adrenal incidentalomas. Eur J Clin Invest 2011;41(5):552–560.
  151. Terzolo M, Pia A, Alì A, Osella G, Reimondo G, Bovio S, Daffara F, Procopio M, Paccotti P, Borretta G, Angeli A. Adrenal incidentaloma: A new cause of the metabolic syndrome? Journal of Clinical Endocrinology and Metabolism 2002;87(3):998–1003.
  152. Morelli V, Reimondo G, Giordano R, Della Casa S, Policola C, Palmieri S, Salcuni AS, Dolci A, Mendola M, Arosio M, Ambrosi B, Scillitani A, Ghigo E, Beck-Peccoz P, Terzolo M, Chiodini I. Long-term follow-up in adrenal incidentalomas: An Italian multicenter study. Journal of Clinical Endocrinology and Metabolism 2014;99(3):827–834.
  153. Deutschbein T, Reimondo G, Di Dalmazi G, Bancos I, Patrova J, Vassiliadi DA, Nekić AB, Debono M, Lardo P, Ceccato F, Petramala L, Prete A, Chiodini I, Ivović M, Pazaitou-Panayiotou K, Alexandraki KI, Hanzu FA, Loli P, Yener S, Langton K, Spyroglou A, Kocjan T, Zacharieva S, Valdés N, Ambroziak U, Suzuki M, Detomas M, Puglisi S, Tucci L, Delivanis DA, Margaritopoulos D, Dusek T, Maggio R, Scaroni C, Concistrè A, Ronchi CL, Altieri B, Mosconi C, Diamantopoulos A, Iñiguez-Ariza NM, Vicennati V, Pia A, Kroiss M, Kaltsas G, Chrisoulidou A, Marina L V., Morelli V, Arlt W, Letizia C, Boscaro M, Stigliano A, Kastelan D, Tsagarakis S, Athimulam S, Pagotto U, Maeder U, Falhammar H, Newell-Price J, Terzolo M, Fassnacht M. Age-dependent and sex-dependent disparity in mortality in patients with adrenal incidentalomas and autonomous cortisol secretion: an international, retrospective, cohort study. Lancet Diabetes Endocrinol 2022;10(7):499–508.
  154. Toniato A, Merante-Boschin I, Opocher G, Pelizzo MR, Schiavi F, Ballotta E. Surgical versus conservative management for subclinical cushing syndrome in adrenal incidentalomas: A prospective randomized study. Ann Surg 2009;249(3):388–391.
  155. Sereg M, Szappanos Á, Tóke J, Karlinger K, Feldman K, Kaszper É, Varga I, Gláz E, Rácz K, Tóth M. Atherosclerotic risk factors and complications in patients with non-functioning adrenal adenomas treated with or without adrenalectomy: A long-term follow-up study. Eur J Endocrinol 2009;160(4):647–655.
  156. Tsuiki M, Tanabe A, Takagi S, Naruse M, Takano K. Cardiovascular risks and their long-term clinical outcome in patients with subclinical Cushing’s syndrome. Endocr J 2008;55(4):737–745.
  157. Chiodini I, Viti R, Coletti F, Guglielmi G, Battista C, Ermetici F, Morelli V, Salcuni A, Carnevale V, Urbano F, Muscarella S, Ambrosi B, Arosio M, Beck-Peccoz P, Scillitani A. Eugonadal male patients with adrenal incidentalomas and subclinical hypercortisolism have increased rate of vertebral fractures. Clin Endocrinol (Oxf) 2009;70(2):208–213.
  158. Tauchmanová L, Pivonello R, De Martino MC, Rusciano A, De Leo M, Ruosi C, Mainolfi C, Lombardi G, Salvatore M, Colao A. Effects of sex steroids on bone in women with subclinical or overt endogenous hypercortisolism. Eur J Endocrinol 2007;157(3):359–366.
  159. Chiodini I, Mascia ML, Muscarella S, Battista C, Minisola S, Arosio M, Santini SA, Guglielmi G, Carnevale V, Scillitani A. Subclinical hypercortisolism among outpatients referred for osteoporosis. Ann Intern Med 2007;147(8):541–548.
  160. Chiodini I, Vainicher CE, Morelli V, Palmieri S, Cairoli E, Salcuni AS, Copetti M, Scillitani A. Endogenous subclinical hypercortisolism and bone: A clinical review. Eur J Endocrinol 2016;175(6):R265–R282.
  161. Guerrieri M, Campagnacci R, Patrizi A, Romiti C, Arnaldi G, Boscaro M. Primary adrenal hypercortisolism: Minimally invasive surgical treatment or medical therapy? A retrospective study with long-term follow-up evaluation. Surg Endosc 2010;24(10):2542–2546.
  162. Peppa M, Boutati E, Koliaki C, Papaefstathiou N, Garoflos E, Economopoulos T, Hadjidakis D, Raptis SA. Insulin resistance and metabolic syndrome in patients with nonfunctioning adrenal incidentalomas: A cause-effect relationship? Metabolism 2010;59(10):1435–1441.
  163. Garrapa GGM, Pantanetti P, Arnaldi G, Mantero F, Faloia E. Body Composition and Metabolic Features in Women with Adrenal Incidentaloma or Cushing’s Syndrome. J Clin Endocrinol Metab 2001;86(11):5301–5306.
  164. Fernández-Real JM, Ricart Engel W, Simó R, Salinas I, Webb SM. Study of glucose tolerance in consecutive patients harbouring incidental adrenal tumours. Clin Endocrinol (Oxf) 1998;49(1):53–61.
  165. Yener S, Genc S, Akinci B, Secil M, Demir T, Comlekci A, Ertilav S, Yesil S. Carotid intima media thickness is increased and associated with morning cortisol in subjects with non-functioning adrenal incidentaloma. Endocrine 2009;35(3):365–370.
  166. Yener S, Baris M, Secil M, Akinci B, Comlekci A, Yesil S. Is there an association between non-functioning adrenal adenoma and endothelial dysfunction? J Endocrinol Invest 2011;34(4):265–270.
  167. Ermetici F, Dall’Asta C, Malavazos AE, Coman C, Morricone L, Montericcio V, Ambrosi B. Echocardiographic alterations in patients with non-functioning adrenal incidentaloma. J Endocrinol Invest 2008;31(6):573–577.
  168. Androulakis II, Kaltsas GA, Kollias GE, Markou AC, Gouli AK, Thomas DA, Alexandraki KI, Papamichael CM, Hadjidakis DJ, Piaditis GP. Patients with apparently nonfunctioning adrenal incidentalomas may be at increased cardiovascular risk due to excessive cortisol secretion. Journal of Clinical Endocrinology and Metabolism 2014;99(8):2754–2762.
  169. Lopez D, Luque-Fernandez MA, Steele A, Adler GK, Turchin A, Vaidya A. “Nonfunctional” Adrenal Tumors and the Risk for Incident Diabetes and Cardiovascular Outcomes. Ann Intern Med 2016;165(8):533.
  170. Elhassan YS, Alahdab F, Prete A, Delivanis DA, Khanna A, Prokop L, Murad MH, O’Reilly MW, Arlt W, Bancos I. Natural History of Adrenal Incidentalomas With and Without Mild Autonomous Cortisol Excess. Ann Intern Med 2019;171(2):107.
  171. Terzolo M, Reimondo G. Insights on the Natural History of Adrenal Incidentalomas. Ann Intern Med 2019;171(2):135.
  172. Iñiguez-Ariza NM, Kohlenberg JD, Delivanis DA, Hartman RP, Dean DS, Thomas MA, Shah MZ, Herndon J, McKenzie TJ, Arlt W, Young WF, Bancos I. Clinical, Biochemical, and Radiological Characteristics of a Single-Center Retrospective Cohort of 705 Large Adrenal Tumors. Mayo Clin Proc Innov Qual Outcomes 2018;2(1):30–39.
  173. Nieman LK, Biller BMK, Findling JW, Murad MH, Newell-Price J, Savage MO, Tabarin A. Treatment of Cushing’s Syndrome: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2015;100(8):2807–2831.
  174. Nieman LK. Approach to the patient with an adrenal incidentaloma. Journal of Clinical Endocrinology and Metabolism 2010;95(9):4106–4113.
  175. Terzolo M, Bovio S, Pia A, Reimondo G, Angeli A. Management of adrenal incidentaloma. Best Pract Res Clin Endocrinol Metab 2009;23(2):233–243.
  176. Calissendorff J, Juhlin CC, Sundin A, Bancos I, Falhammar H. Adrenal myelolipomas. Lancet Diabetes Endocrinol 2021;9(11):767–775.
  177. Di Dalmazi G, Berr CM, Fassnacht M, Beuschlein F, Reincke M. Adrenal Function After Adrenalectomy for Subclinical Hypercortisolism and Cushing’s Syndrome: A Systematic Review of the Literature. J Clin Endocrinol Metab 2014;99(8):2637–2645.
  178. Khawandanah D, ElAsmar N, Arafah BM. Alterations in hypothalamic-pituitary-adrenal function immediately after resection of adrenal adenomas in patients with Cushing’s syndrome and others with incidentalomas and subclinical hypercortisolism. Endocrine 2019;63(1):140–148.
  179. Kastelan D, Kraljevic I, Dusek T, Knezevic N, Solak M, Gardijan B, Kralik M, Poljicanin T, Skoric-Polovina T, Kastelan Z. The clinical course of patients with adrenal incidentaloma: Is it time to reconsider the current recommendations? Eur J Endocrinol 2015;173(2):275–282.
  180. Yeomans H, Calissendorff J, Volpe C, Falhammar H, Mannheimer B. Limited value of long-term biochemical follow-up in patients with adrenal incidentalomas-a retrospective cohort study. BMC Endocr Disord 2015;15(1):6.
  181. Muth A, Taft C, Hammarstedt L, Björneld L, Hellström M, Wängberg B. Patient-reported impacts of a conservative management programme for the clinically inapparent adrenal mass.; 2013:228–236.

 

The Role Of Parents In The Care Of Children With Dyslipidemia

ABSTRACT

 

Parents should be viewed as an integral part of the child’s healthcare team, being both legally and morally responsible for providing proper care to the child. In this paper, we discuss the role of parents as critical members of the healthcare team in caring for youth with dyslipidemia and how clinicians can best leverage this important resource.

 

INTRODUCTION

 

Providing healthcare to a child with a chronic medical condition requires a multidisciplinary team of specially trained and experienced healthcare professionals. Cooperation, empathy, and effective communication between the child, caregivers, and the healthcare team all play key roles in achieving success. This unique model, consistently applied in the healthcare setting, is a cornerstone in promoting physical and emotional wellbeing, and improving outcomes. The development of effective communication takes time and practice, with the goal of developing trust, enhancing bidirectional understanding, and facilitating shared decision-making. Parents should be viewed as an integral part of the child’s healthcare team, being both legally and morally responsible for providing proper care to the child. In this paper, we discuss the role of parents as critical members of the healthcare team in caring for youth with dyslipidemia and how clinicians can best leverage this important resource.

 

THE CLINICIAN-CHILD-PARENT RELATIONSHIP

 

In contrast to adult healthcare, the treatment of children (<18 years-old) is triangulated between the child, parent, and clinician (1). As in all medical encounters, clinicians are provided intimate details about the child and family, based upon perceptions of respect for their autonomy and assurances of confidentiality.

 

Pediatric healthcare professionals routinely consider a child’s age, developmental level, and likes/interests in their clinical interactions and recommendation for care. Their approach is modified as the child grows and matures, building upon a foundation of trust and mutual respect. Yet, given their pivotal role, few clinicians are trained to assess the best way to communicate with the parent based upon the latter’s communication and parenting style. Establishing trust in clinical encounters takes time and a conscious effort by the clinician, and includes getting to know the child and family, providing factual information in a timely manner, use of simple language and examples, and most importantly, the clinician’s willingness to listen. Parents need to feel included and assured that the healthcare team is there to support them in providing for their child’s health and wellbeing. Thus, it is the clinician’s responsibility to find ways to build trust, facilitate effective communication, and identify barriers to success that best serve the needs of the child and their parents.

 

THE “PERSONALIZED “CLINIC NOTE

 

 “Parents don’t care how much you know, until they know how much you care.” High quality healthcare is more than addressing a child’s chief complaint. During the initial clinical encounter, a clinician should strive to get to know the child and the family, developing an understanding of who they are, where they come from, what they do for a living, personal interests, and healthcare beliefs. Inclusion of personal information in a child’s clinic note can provide insight into the social determinants of health that may affect the child’s care and the parent’s resources in providing for their child’s needs. Such information can provide clues as to how best to assist the family and what additional services and resources may be needed (2). The following are two brief examples of a “personalized” clinic note:

 

Eric is a 12-year-old boy who is homeschooled, plays soccer, and has a schnauzer named “Ringo”. His father is a minister, the mother a CPA. There are 2 siblings, one of whom is autistic. Eric was referred by his primary care physician for high cholesterol noted following a routine screening test.

 

Julie is a 16-year-old girl who attends a public school and wants to become a beautician. Her mother is a single parent who works in retail and has 3 other children. Julie is concerned about her weight and has combined dyslipidemia.

 

Personal details included in clinic notes may provide a nonthreatening context to discuss potentially sensitive topics such as diet, physical activity, weight, healthcare beliefs and practices, and potential barriers to achieving goals (3). This information can be invaluable in helping guide the healthcare team’s approach to patient education and treatment.

 

THE ROLE OF THE PARENT

 

In addition to their many roles, parents of children with dyslipidemia have extended responsibilities, including but not limited to:

 

  1. Modeling healthy behaviors.
  2. Educating themselves and their child about the child’s condition.
  3. Overseeing the child’s medical care, including:
    1. Scheduling and attending clinic visits.
    2. Completing laboratory tests and procedures.
    3. Overseeing medication(s), if prescribed.
    4. Helping implement recommendations such as therapeutic lifestyle changes.
    5. Managing healthcare costs.

 

As such, parents play an integral role in the successful outcome of the child with a chronic health condition. By engaging parents in their child’s care, clinicians can increase the likelihood of the child’s compliance with lifestyle changes and treatment recommendations (4).

 

PARENTING STYLES

 

Psychologists suggest that there is a close relationship between a parent’s parenting style and their child’s behavior. Different parenting styles can also contribute to a child’s short- and long-term health outcomes (5).

 

Figure 1. Parenting styles.

 

As clinicians get to know the child and family through clinical interviews, certain questions can be used to gauge a caregiver’s parenting style, which are summarized below.

 

Table 1. Characteristics of Various Parenting Style

AUTHORITATIVE

AUTHORITARIAN

PERMISSIVE

NEGLECTFUL

Warm and Receptive

Unresponsive

Warm/Responsive

Cold/Unresponsive

Clear Rules

Strict Rules

Few or No Rules

No Rules

High Expectations

High Expectations

Indulgent

Uninvolved

Supportive

Value Independence

Expected Blind Obedience

Lenient

Indifferent

 

During a clinic visit, a few simple questions can often provide insight about parenting styles.  For example, you may ask the child:

 

Do you have any household chores? If so, what happens if you fail to do them? The interpretation of the answers is shown in table 2.

 

Table 2. Examples of a Child’s Response Based Upon Parenting Style

AUTHORITATIVE

AUTHORITARIAN

PERMISSIVE

NEGLECTFUL

“Yes”

“Yes”

“Sometimes”

“No”

“My mom helps me.”

“I can’t play video games for a week.”

“I do them if I remember or have time.”

“Nothing."

 

Based on the parenting style, a clinician can determine how best to engage the parent in the child’s care. The following is an example of a common clinical scenario and how caregivers with different parenting styles might respond.

 

Arturo is a 14-year-old boy with familial hypercholesterolemia (FH). He has a confirmed pathologic variant in the low-density lipoprotein (LDL) receptor. He plays the trombone in the school band. His father had a fatal MI at 42 years-of-age; the mother, who has T2D, works as a bank teller and has one other child. His current medications include atorvastatin 20 mg + ezetimibe 10 mg daily.

 

Laboratory test results are shown in table 3.

 

Table 3. Laboratory Test Results

Visit

TC

TG

HDL-C

LDL-C

Visit #1

273

54

59

203

Visit #2

179

81

56

107

Visit #3

159

52

60

89

Visit #4

196

91

43

135

Visit #5

161

82

51

94

Today

220

62

46

162

Goal

<170 mg/dl

<150 mg/dl

>40 mg/DL

<100 ng/dL

 

Based on Arturo’s lab results, the clinician tells the mother, “I am concerned Arturo may have been inconsistent in taking his statin.”

 

A questionnaire, completed independently by both the child and parent prior to the visit, can provide valuable insight into perceptions of compliance. Responses can help guide the clinician’s approach during the visit, addressing concerns about side effects, proper medication administration, financial barriers, and the importance of compliance (Figure 2).

 

Figure 2. Self-Reported Medication Questionnaires

 

When confronting a parent about a child’s inadequate adherence to medical management, the caregiver’s parenting style may dictate their response (figure 3).

 

Figure 3. Example of responses based upon parenting style.

 

THE CLINICIAN’S PERSONALITY

 

A clinician’s personality type helps define their style of communication. One commonly used tool to assess an individual’s personality type is the Myers-Briggs Type Indicator (MBTI), a psychometric questionnaire designed to help understand how people perceive the world and make decisions (figure 4) (6).

 

Figure 4. The Briggs Myers Type Indicator (MBT I).

 

To improve communication and help build trust, it is often useful for clinicians to adapt their style of communication based upon the personality type of the parent. The following is an example of a clinical scenario in which a parent’s personality type may impact their response to a clinician:

Clinician recommendation: “I think your child would benefit from a statin.” Possible parent responses are shown in figure 5.

 

Figure 5. Parent Responses.

 

Knowing the parent’s personality style, a clinician can modify their language to facilitate understanding and help the parent intensify strategies which are likely to be successful.

 

DETERMINANTS OF CLINICAL BEHAVIOR

 

Psychologists have identified two basic dimensions of clinician behavior during a clinic visit.

 

  1. Control - For most clinician's this is the dominant form of behavior, such as frequent interruptions or a louder voice often used to:
    1. Obtain “pertinent information.”
    2. Control the direction and tempo of the interview; and
    3. Stay within the time allocated for the visit.
  2. Affiliation - This behavior reflects friendliness and psychosocial orientation (e.g., showing concern, smiling, offering help).

 

It comes as no surprise that there is a positive association between a clinician’s affiliative behavior and parental perception. But what about clinicians who focus on control? Studies of clinician speech complexity and interruptions have shown that interrupting behavior is negatively associated with recall of medical information and parental satisfaction, especially when used by male clinicians, and that parents report lower satisfaction when clinicians employ more complex language (7).  

 

Some forms of interruption, however, may be perceived as positive, such as when clinicians employ them to enhance understanding, provide assistance, communicate support, or ask for clarification. Here are some examples:

 

“Pardon me for interrupting, [respect] but I want to be clear on what you just shared with me.” [interest, asking for clarification]

 

“I can understand how difficult it must be talking about the loss of your husband. [empathy] If you would like, we can talk about this later”. [concern, compassion]

 

EFFECTIVE CLINICIAN COMMUNICATION

 

Communication during a clinic visit is often facilitated by asking the parents open-ended questions, such as the following.

 

  • What concerns, if any, do you have about your child’s cholesterol?
  • What has been your experience with medications to lower cholesterol?
  • How would you feel about treating your child with medication to lower his/her cholesterol?

 

During follow-up clinic visits, it is often informative to ask children and parents to share what they have learned about their condition at previous visits. For example, ask the child or parent:

 

  1. What they remember about their last clinic visit.
  2. To explain their understanding of cholesterol and triglycerides, and what effects high levels may have on their health.
  3. What medication the child is taking and the proper way to take it.
  4. The likelihood early treatment can prevent heart disease in adulthood.

 

Another way to assess understanding is to ask the child or parent to explain the child’s medical condition and need for medication and monitoring to a medical student or resident present during the clinic visit.

 

Some children and parents may be more comfortable answering theoretical questions or discussing 3rd person examples. For example, you may ask a parent:

 

“Before we talk about your son, John, today, I would appreciate your advice. I saw a 10-year-old boy this morning whose 42-year-old father recently survived a heart attack. Like his father, the son has a very high blood cholesterol level. Having experienced something similar in your family, do you have any advice as to how I can best help this family? What do you feel would be the mother’s main concerns and how should they be addressed?”

 

TRANSITIONAL CARE

 

As they become young adults, the roles and responsibilities of the child verses those of the parents change, necessitating a change by the treating physician.  

 

Children are considered adults when they are 18 years-of-age and older. Unless declared incompetent, they have the legal right to make medical decisions for themselves. At 18, health care providers and clinic staff are not legally permitted to disclose a young adult’s medical information or discuss his/her health status or treatment with anyone - even the parent - although the young adult may still be covered by their parent’s health insurance plan. Thus, at 18, it is the responsibility of the young adult to decide who can be involved in and have information pertaining to their care, as well as whether they consent to treatment. According to the Affordable Care Act (ACA), which expanded health care coverage up to 26 years-of-age, as the primary insurance policy holder, a parent may receive a detailed explanation of benefits (EOB) from private insurers, which includes what doctor(s) the young adult visited, what type(s) of procedure(s) took place, and if specimens were sent to a lab for analysis (8). Therefore, one of the unintended consequences of the ACA is that it provides parents access to their adult child’s health information, if that child is still using their parents’ health insurance, which could inherently violate a young adult’s privacy. Information related to sexual or mental health are sensitive topics in many families, and revealing a young adult’s information regarding sexual or mental healthcare could cause relational issues within a family (9).

 

When planning transition into adult health care, it is helpful to review the family’s knowledge of the child’s diagnosis, key findings (e.g., pre- and post-treatment test results, pertinent family history, treatment goals, and risk enhancers such as lipoprotein(a) and genetic test results), reproductive health, family planning, and genetic transmission. Provide recommendations for appropriate future healthcare, discuss how long prescription refills will be available, and review how to access healthcare records. Discuss the importance of timely follow-up, healthcare costs, health insurance, and legal responsibilities and restrictions. Suggest the young adult/parent investigate the potential benefits of:

 

  • HIPAA waiver - Granting the parents (or another trusted adult) access to their records; and their health care provider permission to talk with the parents and other health care providers about their care.
  • Medical power of attorney - Appoint an individual to make health care decisions on their behalf should they become incapacitated due to serious injury or illness.
  • Durable power of attorney - Enables the parent to handle their child’s financial affairs if they were to become incapacitated.
  • Living will - Specifies personal choices about life-extending medical treatment in the event that a person cannot communicate their wishes themselves.

 

CONCLUSION

 

In partnering with parents, clinicians should always strive to treat them with dignity and respect. Listen to their point of view and consider the family’s values, beliefs, and cultural background when discussing your recommendations, and respect their choices. When sharing information, explain all options, treatments, and results in an informative, unbiased, and timely manner. Encourage and empower the parents to participate in all decisions regarding their child and prepare the young adult to do so in the future. Ultimately, by including parents in their child’s care, clinicians can equip children and their families to optimally manage their chronic medical condition both now and in the future.

 

REFERENCES

 

  1. Tates K, Meeuwesen L. Doctor–parent–child communication. A (re)view of the literature. Soc Sci Med. 2001;52(6):839-851. doi:10.1016/s0277-9536(00)00193-3  
  2. Andermann A. Taking action on the social determinants of health in clinical practice: a framework for health professionals. CMAJ. 2016;188(17-18):E474-E483. doi:10.1503/cmaj.160177  
  3. McBride R. Talking to patients about sensitive topics: Communication and screening techniques for increasing the reliability of patient self-report. MedEdPORTAL. Published online 2012. doi:10.15766/mep_2374-8265.9089  
  4. Dalton WT 3rd, Kitzmann KM. Broadening parental involvement in family-based interventions for pediatric overweight: implications from family systems and child health. Fam Community Health. 2008;31(4):259-268. doi:10.1097/01.FCH.0000336089.37280.f8  
  5. Park H, Walton-Moss B. Parenting style, parenting stress, and childrenʼs health-related behaviors. J Dev Behav Pediatr. 2012;33(6):495-503. doi:10.1097/dbp.0b013e318258bdb8  
  6. Woods RA, Hill PB. Myers Brigg. Published online 2023. Accessed September 5, 2023. https://pubmed.ncbi.nlm.nih.gov/32119483/  
  7. Gemmiti M, Hamed S, Wildhaber J, Pharisa C, Klumb PL. Physicians’ speech complexity and interrupting behavior in pediatric consultations. Health Commun. 2022;37(6):748-759. doi:10.1080/10410236.2020.1868063  
  8. Read the Affordable Care Act. Healthcare.gov. Accessed September 5, 2023. https://www.healthcare.gov/where-can-i-read-the-affordable-care-act/  
  9. Campbell-Salome G. “Yes they have the right to know, but…”: Young Adult Women Managing Private Health Information as Dependents. Health Commun. 2019;34(9):1010-1020. doi:10.1080/10410236.2018.1452092

Primary Generalized Glucocorticoid Resistance Syndrome

ABSTRACT

 

Primary generalized glucocorticoid resistance syndrome is a rare genetic disorder characterized by resistance of entire tissues to glucocorticoids. Affected subjects demonstrate elevation of serum cortisol without Cushingoid manifestations, as the hypothalamic-pituitary-adrenal (HPA) axis is upregulated to compensate for the reduced action of this steroid in local tissues. Instead, these patients develop hypertension and/or signs of hyperandrogenism, because hyper-secreted adrenocorticotropic hormone (ACTH) stimulates production of adrenal mineralocorticoids and/or androgens in addition to the glucocorticoid cortisol. At the molecular level, this syndrome is caused by inactivating mutations in the NR3C1 gene that encodes the human glucocorticoid receptor (hGR) protein. Biochemical, molecular and structural exploration on pathologic mutant receptors revealed a variety of functional defects, such as reduced affinity to glucocorticoids or target DNA, inability to transactivate glucocorticoid-responsive genes, and slowing of the cytoplasmic to nuclear translocation. The clinical spectrum of this syndrome is thus broad, ranging from asymptomatic to severe cases of mineralocorticoid and/or androgen excess depending on the severity of genetic defects and resulting dysfunction of the mutated receptors. When this syndrome is suspected, a detailed personal and family history should be obtained. Physical examination should include an assessment for signs of mineralocorticoid and/or androgen excess. In neonates and young children, severe hypoglycemia and loss of consciousness due to reduced actions of glucocorticoids in the liver may be present as initial manifestations in addition to hypertension and/or genital abnormalities. Suspected subjects should undergo a detailed endocrinologic evaluation with particular emphasis on the measurement of diurnal serum cortisol and plasma ACTH concentrations and determination of the 24-hour urinary free cortisol excretion to identify upregulation of the HPA axis with preservation of the normal circadian rhythmicity. The diagnosis of this syndrome should be confirmed by sequencing of the NR3C1 gene including exon/intron junctions and subsequent validation of functional defects of the mutated receptors. Treatment involves administration of high doses of mineralocorticoid activity-sparing pure glucocorticoids like dexamethasone, which stimulate the mutant and/or the wild-type hGR, and suppress the endogenous secretion of ACTH and adrenal steroids in the affected subjects.

 

INTRODUCTION

 

Organisms are exposed continuously to internal and external stressors, and live through them by maintaining the internal equilibrium called homeostasis (1). In order to respond adequately to such stressors through coordinating various body activities, we humans are equipped with a highly sophisticated stress responsive system, the hypothalamic-pituitary-adrenal (HPA) axis, which consists of the brain hypothalamus, the anterior pituitary gland, and the adrenal cortex, and employs glucocorticoids as its end-effector hormones. Actions of glucocorticoids, which are essential for life, can be determined by a balance between circulating levels of these hormones and local tissue sensitivity (2, 3). Exceeding appropriate ranges of tissue sensitivity to glucocorticoids may present either as glucocorticoid resistance or glucocorticoid hypersensitivity with their specific manifestations (3, 4). Such alterations in tissue glucocorticoid actions can occur in general (that is, throughout the body) or in tissue-specific manner (restricted in some organs and tissues; e.g., immune organs/cells, central nervous system (CNS), liver and fat tissues) (3). They are caused primarily by genetic defects of the molecules involved in the glucocorticoid signaling pathway or secondary through modulation of this pathway by other pathologic conditions, such as infectious, inflammatory and autoimmune diseases, obesity, and insulin resistance/overt diabetes mellitus. One such condition is the primary generalized glucocorticoid resistance syndrome, which is caused by inactivating mutations in the glucocorticoid receptor gene (5). Affected subjects develop partial glucocorticoid resistance observed in entire organs and tissues of the affected subjects (5). In recognition of Professor George P. Chrousos' novel and extensive research work in this field, the term “Chrousos Syndrome” may be used for this syndrome (6, 7).

 

GLUCOCORTICOIDS

 

Glucocorticoids (cortisol in humans and corticosterone in rodents) are produced from cholesterol through multiple enzymatic reactions in the zona fasciculata of the adrenal cortex in response to the adrenocorticotropic hormone (ACTH) released from the pituitary gland (1). Glucocorticoids regulate a broad spectrum of physiologic functions essential for life, such as growth, reproduction, immunity, intermediary metabolism, cardiovascular tone, and CNS functions, playing essential and indispensable roles in the maintenance of resting and stress-related homeostasis (1, 7, 8). In addition, glucocorticoids exert potent anti-inflammatory and immunomodulatory effects particularly with their stress-equivalent or pharmacologic doses, thus they are widely used in the treatment of inflammatory, autoimmune, and lymphoproliferative diseases (8).

 

GLUCOCORTICOID RECEPTOR PROTEINS, ISOFORMS AND ITS ENCODING GENE, NR3C1

 

Circulating cortisol freely passes through the cytoplasmic membrane and enters into the cytoplasm of its target cells, and binds to an intracellular protein, the glucocorticoid receptor (GR) (9, 10). The human (h) GR is one of the steroid/thyroid/retinoic acid nuclear hormone receptor superfamily proteins, which consist of over 600 members in the animal kingdom (11). Many of them mediate extracellular signals transduced mainly by lipophilic hormones/compounds into the cell nucleus by binding them as ligands and by acting as ligand-dependent transcription factors (12, 13). hGR influences transcription rates of numerous glucocorticoid-responsive genes (up to 3~5% of the entire protein-coding genes) in a positive or a negative fashion by interacting directly or indirectly with promoter/enhancer regions of these genes (14). The hGR gene (NR3C1: nuclear receptor subfamily 3, group C, member 1) consists of 9 exons and is located at chromosome 5q31.3. Exons 2-9 constitute the protein-coding sequence, whereas exon 1 encodes an untranslated region (12, 14, 15). The human NR3C1 gene has multiple exon 1s (see below) that harbor specific promoters containing a respective transcription start site for conferring tissue-specific expression of the receptor protein (15). Alternative splicing of the NR3C1 gene in exon 9s generates two highly homologous receptor isoforms, the hGRα and the hGRβ (16). They share amino (N)-terminal 727 common amino acids, but then diverge, with hGRα having an additional 50 amino acids and hGRβ having an additional, nonhomologous 15 amino acids at their carboxyl (C)-termini (17). hGRα resides primarily in the cytoplasm of cells and represents the classic GR that binds natural and synthetic glucocorticoids and mediates most of the actions of these hormones (15). On the other hand, hGRβ does not bind glucocorticoids, has intrinsic, gene-specific transcriptional activity, and exerts a dominant negative effect on the transcriptional activity of hGRa (18). Although physiologic and pathologic roles of hGRβ are still largely unknown (19, 20), recent studies demonstrated that this isoform is implicated in modulation of the insulin signaling and participates in the pathogenesis of brain gliomas (21-23).  

 

The hGRα mRNA expresses not only the classic, full-length hGRα, but also multiple translational isoforms by using at least eight alternative amino-terminal translation initiation sites (24). All these hGRα isoforms are differentially distributed in the cytoplasm and/or the nucleus in the absence of ligand, have different transcriptional activity, and display distinct transactivating or transrepressing activities on various glucocorticoid-responsive genes (24). Since hGRβ shares with hGRα a common amino-terminal domain that contains the same translation initiation sites, the hGRβ variant mRNA might also be translated through the same translation initiation sites to a similar host of hGRβ isoforms (14).

 

The human NR3C1 has 11 different promoters with their alternative first exons (1A1, 1A2, 1A3, 1B, 1C, 1D, 1E, 1F, 1H, 1I and 1J) (25, 26). Therefore, it can produce 11 different hGRa mRNA transcripts from different promoters that encode the same hGRa protein, as these transcripts share common exon 2 to exon 9a that contains the same translation initiation codon. 1A1, 1A2, 1A3 and 1I are located in the distal promoter region spanning ~32,000-36,000 bps upstream of the translation initiation site, while 1B, 1C, 1D, 1E, 1F, 1H and 1J position in the proximal promoter region located up to ~5,000 bps upstream of this site (25). Through differential use of these promoters, expression levels of hGRa can vary among tissues in different physiologic and pathologic conditions, as each tissue has specific expression profiles of local transcription factors and epigenetic modification of chromatin-associated molecules bound on these exon 1-associated promoters (25, 27, 28). Again, differential tissue-specific expression of hGRb through the use of these promoters appears to be present. The above-indicated marked complexity in transcription/translation of the human NR3C1 gene enables target tissues to respond differently to circulating cortisol and accounts for stochastic, but still a highly organized nature of tissue glucocorticoid actions, in order to fulfil specific local needs of glucocorticoid hormonal effects (14). Such complexity of the glucocorticoid signaling at the receptor level also indicates that the proper biologic action of glucocorticoids in every target tissue is extremely important.

 

The hGRα protein consists of three major domains and one region, namely the N-terminal (NTD), DNA-binding (DBD) and ligand-binding domain (LBD), and the hinge region (HR) (15). Exact amino acid location of these domains/region in the hGRa protein explained below is based on the data retrieved from the Pfam source of the Ensembl database (www.ensembl.org). NTD is encoded by exon 2 and represents the largest domain of the receptor, spanning over amino acids 1 to 401. It contains an unstructured acidic transactivation surface called activation function (AF) -1, which is used as a molecular platform for modulating the transcription of glucocorticoid-responsive genes (10). This domain also undergoes several post-translational modifications particularly at AF-1.  DBD is expressed from exons 3 and 4, and lies between amino acids 417 and 494. This domain consists of two 4C (cysteine)-type zinc fingers and support the interaction between the receptor and its target DNA sequences known as glucocorticoid response elements (GREs) (10, 29). LBD is encoded by exons 5-9 and positions at the C-terminal end of the receptor corresponding to amino acids 531 to 777. This domain is structurally formed with 12 a-helices and four b-sheets, and contains two functional structures, the ligand-binding pocket (LBP) and the second transactivation surface called AF-2, as well as several other molecular platforms including the one responsible for nuclear translocation of the receptor (29, 30). Most of the protein surfaces of LBD that mediate these LBD-specific functions are formed upon binding of the receptor to a ligand and following conformational changes of this domain (15). Finally, HR lies between DBD and LBD, is encoded by 5’ part of exon 5, and spans between amino acids 495 and 530. This region provides appropriate structural flexibility to the receptor and allows the dimerized receptors to interact with different classic/alternative tandem GREs with various length of spacing nucleotides (the classic tandem GREs has three spacing nucleotides)  (15).

 

MOLECULAR ACTIONS OF hGRa

 

Intracellular Shuttling of hGRa and its Regulators

 

At target cells, hGRα in the absence of glucocorticoids resides primarily in the cytoplasm as part of the hetero-oligomeric complex consisting of chaperone heat shock proteins (HSPs) 90, 70 and 50, immunophilins (e.g., FK506-binding protein (FKBP)), and possibly other proteins (31) (Figure 1). Binding of HSP90 to hGRα induces a conformational change in receptor’s LBD, and confers its ligand-friendly state, exposing the LBP to glucocorticoids and masking two nuclear localization signals (NLS), NL1 and NL2. Upon binding to a ligand, hGRα dissociates from the complex, exposes NL1 and NL2 to their counterpart molecular machinery, and translocates into the nucleus through the nuclear pore. NL1 harbors a classic NLS and is located between the C-terminal portion of DBD and the N-terminal part of HR (32). The function of NL1 is dependent on the importin a, a protein component of the nuclear pore-associated nuclear import system, which transports a liganded GRa as a cargo from the cytoplasm to the nucleus through the nuclear pore in an ATP-dependent fashion (33). NL2 spans over most of the LBD whose molecular mechanism(s) for supporting nuclear translocation of the receptor has(ve) not yet been elucidated (31, 34). Inside the nucleus, ligand-bound hGRα dimerizes and modulates transcription rates of glucocorticoid-responsive genes by associating with promoter/enhancer regions of their encoding genes (15) (Figure 1). The receptor subsequently liberates the ligand and is dissociated from its target genes and slowly translocates back to the cytoplasm with the molecular mechanisms described below (15). The ubiquitin-proteasomal pathway degrades some of the liganded hGRa in the nucleus, facilitating clearance of the receptor from GREs; thus this system negatively regulates the transcriptional activity of hGRa (35).

 

In addition to translocating into the nucleus, some liganded hGRas migrate to the cytoplasmic membrane where they modulate the activity of cell surface receptors by associating with their intracellular signaling molecules, such as classic and small GTP-binding (G) proteins, and several serine/threonine and tyrosine kinases (36-38). The ligand-bound hGRais also known to translocate into the mitochondria and to modulate the activity of this intracellular organelle (39). After modulating transcription rates of glucocorticoid-responsive genes in the nucleus, ligand-liberated hGRα is exported back to the cytoplasm and is re-incorporated into the HSP-containing multiprotein complex to function again as a ligand-binding competent receptor (31, 40) (Figure 1). Several mechanisms are postulated for mediating the GRa export from the nucleus to the cytoplasm. The Ca2+-binding protein calreticulin plays a role in this process, directly binding to DBD of the receptor (41-43). The chromosomal maintenance 1 (CRM1, also known as exportin 1)- and the classic nuclear export signal (NES)-mediated nuclear export machinery does not appear to function directly on hGRa (32, 42). Rather, NES-harboring and phospho-serine/threonine-binding proteins 14-3-3s can bind hGRa, and shift its intracellular localization toward the cytoplasm (44, 45). This action of 14-3-3s on hGRa appears to be independent to the ligand-induced nuclear translocation of the receptor, which is mediated in part by the NL1/importin a-associated nuclear pore complex. Numbers of serine and threonine residues of hGRa are phosphorylated by several serine/threonine kinases at their specific target residues, some of which function as phosphorylation-dependent binding sites of 14-3-3 proteins (46). For example, the v-akt murine thymoma viral oncogene homolog 1 (AKT1) (or the protein kinase B a) phosphorylates serine (S) 134 of the hGRa, and 14-3-3 binds to phosphorylated S134. Binding of 14-3-3 on hGRa at this site shifts subcellular localization of the latter to the cytoplasm and downregulates its transcriptional activity inside the nucleus (45, 47). The misshapen-like kinase 1 (MINK1) and the Rho-associated protein kinase (ROCK) respectively phosphorylate threonine (T) 524 and S617 (48). 14-3-3s bind phosphorylated forms of these residues as a dimer (48), possibly modulating subcellular localization and transcriptional activity of the hGRa.

 

Figure 1. Intracellular circulation and actions of hGRα. hGRα resides in the cytoplasm in the absence of ligand by forming a heterocomplex with several heat shock proteins (HSPs), immunophilins (e.g., FKBP), and some other proteins. Upon binding to ligand cortisol, hGRα dissociates from the complex and translocates into the nucleus through the nuclear pore. Inside the nucleus, hGRα binds directly to glucocorticoid response elements (GREs) located in promoter/enhancer regions of glucocorticoid-responsive genes. DNA-bound hGRα then stimulates transcription rates of glucocorticoid-responsive genes by attracting the regulatory regions the transcription regulatory complex including the RNA polymerase II (RNPII) and its ancillary components through bridging coactivators, such as p300/CBP and p160 proteins. Promoter/enhancer-bound hGRα also recruits in collaboration with these coactivators various chromatin remodeling molecules, including the DRIP/TRAP complex (DRIP/TRAP), the SWI/SNF chromatin modulator (SWI/SNF), and the Mediator complex (MED). In addition to binding directly to DNA and regulating transcription, hGRα interacts indirectly with regulatory regions of glucocorticoid-responsive genes via protein-protein interaction with other transcription factors (TFs) and/or attracted cofactor molecules, ultimately modulating positively and negatively the transcriptional activity of GRE- and non-GRE-containing glucocorticoid-responsive genes. hGRα then moves back to the cytoplasm to re-form a heterocomplex with HSPs for regaining a ligand-friendly status or is cleared from DNA by proteasomal degradation. Further, hGRα can influence the action of cell surface receptors by associating with their intracellular signaling molecules, such as classic and small G-proteins, and several serine/threonine and tyrosine kinases (known as non-genomic actions of glucocorticoids). Accumulating evidence suggests that liganded hGRα also influences the transcription of mitochondrial genes by translocating into this intracellular organelle. CBP: cAMP-responsive element-binding protein (CREB)-binding protein; DRIP/TRAP: vitamin D receptor-interacting protein/thyroid hormone receptor-associated protein complex; FKBPs: FK506-binding proteins; GREs: glucocorticoid response elements; GR: glucocorticoid receptor; HSPs: heat shock proteins; MED: Mediator complex; p160: p160-type nuclear receptor coactivator; RNPII: RNA polymerase II; SWI/SNF: switching/sucrose non-fermenting complex; TFs: transcription factors; TREs: transcription factor response elements.

 

Genomic and Non-genomic Actions of hGRα

 

After binding to glucocorticoids and translocating into the nucleus, hGRα binds as a dimer to a tandem GREs located in promoter/enhancer regions of glucocorticoid-responsive genes, and regulates their mRNA expression positively or negatively, depending on the GRE sequence and the promoter/enhancer context (15, 49, 50) (Figure 1). GRE-bound hGRα stimulates transcription of responsive genes by facilitating formation of the transcription regulatory complex, which includes the RNA polymerase II (RNPII) and its ancillary components (51). Mechanically, hGRα uses its two transactivation domains, AF-1 and AF-2, as protein surfaces for interacting with and attracting nuclear receptor coactivators (51). These proteins then act as bridges between the DNA-bound hGRα and the RNPII-containing transcription initiation complex (52, 53) (Figure 1). In addition, they act in themselves as histone acetyltransferases (HAT) as well as attract other enzymatic proteins, and loosen tightly packed chromatin DNA by chemically modulating specific amino acid residues of histones and other chromatin-associated molecules (54). Representatives of these HAT coactivators include p300 and its homologous cAMP-responsive element-binding protein (CREB)-binding protein (CBP), and the p160 family of nuclear receptor coactivators (NCoAs). The former proteins serve as macromolecular docking “platforms” for many transcription factors, including nuclear hormone receptors, CREB, activator protein-1 (AP-1), nuclear factor-κB (NF-κB), p53, and signal transducers and activators of transcription (STATs), and thus, are called co-integrators (55). On the other hand, the p160 family of nuclear receptor coactivators (NCoAs) is more specific to nuclear hormone receptors including hGRa, and play a central role in the initiation of transcription by hGRa, as they are first attracted to the DNA-bound receptor molecule (55, 56). For physical interaction with hGRa, p160-type coactivators employ the LxxLL motif in which “L” is leucine and “x” is any amino acids. They harbor in their nuclear receptor-binding domain (NRB) multiple LxxLL motifs, each of which have different affinity to respective nuclear hormone receptors (55-58). The LxxLL motif forms the a-helical structure and is deeply buried into the molecular cleft formed by the AF-2 surface of the liganded hGRa (58). Interestingly, p160 family proteins also serve as transcriptional coactivators for some other transcription factors (e.g., NF-kB) (59, 60). In collaboration with these transcriptional coactivators and promoter/enhancer-bound other transcription factors, hGRα interacts with and attracts several distinct chromatin remodeling complexes (e.g., the mating-type switching/sucrose non-fermenting (SWI/SNF) complex, the vitamin D receptor-interacting protein/thyroid hormone receptor-associated protein (DRIP/TRAP) complex, and the Mediator (MED) complex) as well as various enzymatic molecules, scaffold proteins, and long non-coding RNAs (e.g., the steroid receptor RNA coactivator (SRA) and the growth arrest-specific 5 (Gas5)), ultimately forming a huge transcriptional regulatory complex for initiating transcription of the downstream coding sequence though the attracted RNPII (61-64). These newly identified functional oligonucleotides exert their transcriptional regulatory activity in part by modulating the liquid-liquid phase separation among various proteins inside the transcription regulatory complex formed on the DNA-bound hGRa (65).

 

Similar to the transcription factors incorporated in the transcriptional regulatory complex recruited by GREs-bound hGRα, liganded hGRα is also attracted to the transcription regulatory complex formed by DNA-bound other transcription factors (e.g., AP-1, NF-κB, p53, STATs, and forkhead transcription factors: FOXOs). This incorporation of hGRα can be independent to its physical association with DNA GREs, and the recruited hGRα modulates their transcriptional activity positively or negatively (15, 66) (Figure 1). The interaction between hGRα and these transcription factors are mediated by mutual protein-protein interactions between these proteins or indirectly through bridging coactivators, such as p300/CBP and p160-type coactivators (15). This GRE-independent activity of hGRα may be more important than the GRE-mediated one, given that the mice harboring a mutant GR defective in the dimerization surface, and thus, active in protein-protein interaction but inactive in transactivation via tandem GREs, survive and procreate, in contrast to the mice with Nr3c1 gene knock-out, which die immediately after birth due to respiratory failure (67). Suppression of transactivation of other transcription factors through such protein-protein interactions appears to be important particularly in the suppression of immune functions and inflammation by glucocorticoids (68-70).

 

Mounting evidence suggests that glucocorticoids also signal within seconds or minutes. These effects are called “non-genomic”, since they do not require the transcriptional activity of hGRα (15). Representative examples of these actions are: (i) the immediate suppression of ACTH release from the anterior pituitary gland by glucocorticoids (71); (ii) the increased frequency of excitatory post-synaptic potentials by glucocorticoids in the brain hippocampus (72); (iii) the cardioprotective role of glucocorticoids through nitric oxide-mediated vasorelaxation (73); and (iv) some immunomodulatory effects of glucocorticoids via inhibition of the T-cell receptor signaling (74). Some of the molecular mechanisms underlying these actions of hGRα have been proposed. For example, ligand-activated hGRα physically interacts with the classic G protein b through its NTD, and may modulate the action of G protein-coupled receptors located at the cytoplasmic membrane (36). Recent studies also demonstrated that hGRα influences the activity of kinase-mediated signaling, such as of the mitogen-activated protein kinase and the phosphatidylinositol 3-kinase through interacting with their key signaling molecules residing under the cytoplasmic membrane or in the cytoplasm (71-75)(Figure 1).

 

These non-genomic effects of hGRα modulate the action of some intracellular signaling pathways, whereas the latter can influence the activity of hGRα through post-translational modifications (PTMs) of this receptor protein. Such PTMs include phosphorylation, ubiquitination, acetylation, and sumoylation (15). These covalent changes may influence receptor stability, subcellular localization, as well as its interaction with other proteins including transcription factors and transcriptional cofactors/regulators (10). Thus, enzymes catalyzing these PTMs act as molecular effectors of their upstream intracellular signaling pathways for modulating the biologic effects of glucocorticoids by targeting the hGRaprotein.

 

In addition to the above-explained diverse actions, glucocorticoids can modulate expression of the mitochondrial genes by translocating into this cytoplasmic organelle, and by binding to the classic GREs located in some regulatory sites (D-loop) of these genes (76-78) (Figure 1). This action of hGRα in the mitochondria appears to play a role in the glucocorticoid-mediated modulation of apoptosis, a well-known process of the programmed cell death, and may contribute to the therapeutic effects of glucocorticoids on hematologic and other malignancies (79).

 

PRIMARY GENERALIZED GLUCOCORTICOID RESISTANCE SYNDROME

 

Pathophysiology and Clinical Manifestations

 

This syndrome is a condition first described by Chrousos, et.al., as a rare, familial or sporadic, genetic disorder characterized by generalized, partial target tissue insensitivity to glucocorticoids (80). Because of glucocorticoid insensitivity in the central components of the HPA axis, glucocorticoid-mediated negative feedback inhibition on the brain hypothalamus and the anterior pituitary gland is decreased (5, 81) (Figure 2). These changes result in compensatory elevation of the corticotropin-releasing hormone (CRH) and the arginine-vasopressin (AVP) at the hypothalamus and systemic release of the ACTH from the anterior pituitary gland. Excess ACTH secretion then causes bilateral adrenocortical hyperplasia and increased production/secretion of cortisol, which compensates for its reduced actions in target tissues. However, elevated circulating ACTH also stimulates production of other adrenal steroids, such as mineralocorticoids (e.g., deoxycorticosterone (DOC) and corticosterone) and/or adrenal androgens (e.g., androstenedione, dehydroepiandrosterone (DHEA), and DHEA-sulfate (DHEA-S)), leading to the development of excess manifestations of these hormones, because tissue sensitivity to these steroids is not altered. Increased mineralocorticoids may cause hypertension and/or hypokalemic alkalosis, whereas elevated adrenal androgens may develop manifestations (see below) through their direct effects on target tissues and/or indirect actions via modulation of the hypothalamic-pituitary-gonadal axis.

 

Figure 2. Pathophysiologic mechanisms and clinical manifestations of primary generalized glucocorticoid resistance syndrome (PGGRS). The HPA axis consists of the brain hypothalamus, the anterior pituitary gland, and the adrenal cortex with their secreting hormones/peptides, CRH/AVP, ACTH and cortisol, respectively. In patients with this syndrome, their HPA axis is re-set to upward with preservation of circadian rhythmicity due to generalized, partial insensitivity to glucocorticoids in entire tissues. Thus, hypothalamic CRH/AVP, pituitary ACTH and adrenal cortisol are all hyper-secreted in order to compensate for the reduced actions of cortisol in both CNS and peripheral tissues. In addition to augmenting production of cortisol in the adrenal glands, elevated ACTH stimulates secretion of mineralocorticoids (e.g., deoxycorticosterone and corticosterone) and androgens (e.g., androstenedione, dehydroepiandrosterone (DHEA) and DHEA-sulfate(S)), which in turn cause a variety of manifestations associated with excess secretion of these hormones. In contrast, manifestations associated with overproduction of cortisol are rare in adult patients but neonates/young children may develop hypoglycemia and associated seizures due to reduced actions of cortisol in the liver. Elevated CRH/AVP in CNS may precipitate anxiety and depression in some patients. Solid lines indicate positive effects, whereas dashed lines show negative effects. Manifestations associated with elevation of the indicated molecules/compounds are shown with red letters. ACTH: adrenocorticotropic hormone; AVP: arginine vasopressin; CNS: central nervous system; CRH: corticotropin-releasing hormone; DHEA: dehydroepiandrosterone; DHEA-S: DHEA-sulfate; PGGRS; primary generalized glucocorticoid resistance syndrome.

 

Manifestations associated with excess adrenal androgens observed in patients with this syndrome include acne, hirsutism (more common in females), decreased fertility in both sexes, male-pattern hair loss, menstrual irregularities and oligo-anovulation in females, and oligospermia in males. Affected children may develop advanced bone age and subsequent short stature in their adulthood. In female new born babies, clitoromegaly/ambiguous genitalia may be seen (5, 82, 83).

 

Clinical manifestations of glucocorticoid deficiency are rare in adult patients but are reported in neonates/young children as severe hypoglycemia and associated seizures/coma, because gluconeogenesis depends on the proper action of glucocorticoids in the liver during early childhood (84-86). Some adult patients develop anxiety and/or chronic fatigue, which appear to be caused by elevated hypothalamic CRH and/or AVP (87-92). Increased circulating ACTH may cause bilateral adrenal hyperplasia (5). Some patients harbor adrenal incidentalomas (93, 94). Although this adrenal neoplasm is very common in general population (95), elevated circulating ACTH may facilitate tumor development and/or its growth. Further, one patient with this syndrome harbored an ACTH-producing pituitary adenoma, which might have been caused/facilitated by the elevated CRH/AVP (96).

 

Finally, the clinical spectrum of this syndrome is broad, ranging from severe to mild forms, and a number of patients may even be asymptomatic, displaying biochemical alterations only (5, 93, 97, 98). This heterogeneity is mainly due to variable impact of the patients’ genetic changes in the receptor protein, but other factors, such as their genetic backgrounds and/or epigenetic and biochemical changes, for example, associated with their ageing and lifestyles, may also contribute to variability of disease expression.

 

NR3C1 Gene Mutations That Cause Primary Generalized Glucocorticoid Resistance Syndrome

 

The molecular basis of this syndrome is ascribed to inactivating mutations in the NR3C1 gene, which impair molecular actions of hGRα and hence decrease tissue sensitivity to glucocorticoids. Currently, 36 pathologic mutations that cause this syndrome have been reported (Table 1 and Figure 3). Chrousos, et. al., reported the first family of this syndrome who carried a homozygous miss-sense mutation, which replaces adenine by thymine at nucleotide position 1,922 (80). The NR3C1 gene harboring this mutation expresses the hGRαD641V mutant receptor, which has valine (V) instead of aspartic acid (D) at amino acid position 641 in the LBD (80). Since then, numbers of patients were reported whose pathologic mutations were identified mostly as heterozygous in the coding sequence of LBD (90, 96, 99-102). Most of these patients demonstrated characteristic manifestations, such as those of mineralocorticoid and androgen excess, similar to the original case of Chrousos, et. al., thus they may be considered as “classic cases”. More recently, technological progress in the genome sequencing including the use of capillary or high through-put next generation sequencers enabled clinical researchers to conduct large studies with recruitment of the subjects with conventional/unconventional manifestations, (e.g., obesity and bilateral adrenal incidentalomas, as evident in the French Muta-GR study (ClinicalTrials.gov Identifier: NCT02810496) (97)). Clinicians are now able to obtain much easier and faster than before the data of patients’ genome sequence around the NR3C1 gene. Together with growing acknowledgement of this syndrome among clinicians and clinical researchers, such technological progress appears to have facilitated the discovery of new cases with classic symptoms, as well as those with much milder and/or alternative manifestations or even with biochemical changes only. Further, the identified mutations tend to distribute over the entire NR3C1 gene including coding areas of all three major domains and intronic sequences (Table 1 and Figure3).

 

Among 36 pathologic NR3C1 mutations, only three are homozygous mutations, while the other 33 are heterozygous (Table 1 and Figure 3). One patient harbors two different NR3C1 mutations each of which are identified in different alleles (thus, compound heterozygous) (86). Among 34 mutations found in the NR3C1 coding sequence, 24 are miss-sense mutations, which replace one amino acid with another (thus, point mutations), five are non-sense mutations, which introduce a stop codon and generate truncated receptor proteins, and another five are frame-shift mutations, which also develop truncated receptors but with additional unrelated amino acids after the mutation point. At the receptor protein level, 22 mutations are located in LBD, two in HR, seven in DBD, and four are in NTD (Figure 3). In addition to these coding sequence mutations, two mutations are identified in the intronic sequence, located in intron F (between exon 5 and 6) and in intron I (between exon 7 and 8), respectively (91, 103).

 

 

Table 1. The NR3C1 Gene Mutations that Cause Primary
Generalized Glucocorticoid Resistance Syndrome

Amino Acid Change

Nucleotide Change

Zygosity

Mutation Type

Proband’s Gender and Age

Clinical Manifestations

Molecular Defects

References

NTD Mutations

P9R

26C>G

Heterozygous

Point Mutation

M, 33

Hypertension

N.D.

(104)

Q123X

367G>T

Heterozygous

Point Mutation

F, 31

Fatigue, Anxiety, Hirsutism, Irregular menstruation, Infertility

N.D.

(87)

E198X

592G>T

Compound heterozygous with 2141G>Amutation

Point Mutation

F, 3

Hypoglycemia

Hypertension

Also harbors R714Q expressed from a different allele

(86)

D401H

1201G>T

Heterozygous

Point Mutation

F, 43

Hypertension

Hyperglycemia

Increased transcriptional activity

(105)

DBD Mutations

V423A

1268T>C

Heterozygous

Point Mutation

M, 9

Fatigue

Anxiety

Hypertension

Decreased DNA-binding activity

(88)

R469X

1405C>T

Heterozygous

Point Mutation

M, 46

Adrenal hyperplasia

Hypertension

Hypokalemia

No GR mRNA and protein expression from the affected allele

(106)

R477C

1429C>T

Heterozygous

Point Mutation

F, 12

Mild hirsutism

Elevated cortisol

N.D.

(92)

R477H

1430G>A

Heterozygous

Point Mutation

F, 41

Hypertension, Hirsutism,

Fatigue

No DNA-binding activity

(107)

R477S

1429C>A

Heterozygous

Point Mutation

F, 30

Hypertension

Elevated serum cortisol

No DNA-binding activity

(93)

Y478C

1433A>G

Heterozygous

Point Mutation

M, 49

 

Adrenal incidentaloma

No symptoms

Decreased DNA-binding activity

(93)

HR Mutations

R491X

1471C>T

Heterozygous

Point Mutation

M, 44

Bilateral adrenal hyperplasia

Elevation of ACTH and cortisol

Decreased transcriptional activity

(97)

Q501H

1503G>T

Heterozygous

Point Mutation

F, 60

No symptoms

Mild elevation of urinary free cortisol

Decreased transcriptional activity

(97)

LBD Mutations

S551Y

1652C>A

Heterozygous

Point Mutation

M, 14

Fatigue

Hypokalemia Hypertension

Polyuria

Decreased affinity to ligand

Decreased transcriptional activity

(108)

T556I

1667C>T

Heterozygous

Point Mutation

M, 56

Adrenal incidentaloma

Increased UFC

N.D.

(94)

I559N

1676T>A

Heterozygous

Point Mutation

M, 33

Hypertension,

Oligospermia, Infertility

No ligand-binding activity

(96, 99)

V571A

1724T>C

Heterozygous

Point Mutation

 

F, 9

Ambiguous genitalia*, Hypertension, Hypokalemic Alkalosis

Hyperandrogenism

Highly decreased ligand-binding activity

(82, 100)

V575G

1724T>G

Heterozygous

Point Mutation

M, 70

Bilateral adrenal hyperplasia

(His daughters have mild hirsutism)

Decreased affinity to ligand

Decreased transcriptional activity

(98)

H588LfsX5

1762-1765insTTAC>G

Heterozygous

Frame Shift

F, 41

Hirsutism

Anxiety

Fatigue

N.D.

(92)

L595V

1915C>G

Heterozygous

Point Mutation

F, 16

No symptoms

Decreased affinity to ligand

Decreased transcriptional activity

(98)

S612YfsX15

1835delC

Heterozygous

Frame Shift

F, 20

Fatigue

Hirsutism

No ligand-binding activity

(109)

D641V

1922A>T

Homozygous

Point Mutation

M, 48

Hypertension, Hypokalemic alkalosis

Reduced affinity to ligand

Reduced transcriptional activity

(80)

Y660X

1992A>T

Heterozygous

Point Mutation

F, 70

Hypokalemia

Hypertension

No transcription activity

(110)

L672P

 

2015T>C

Heterozygous

Point Mutation

M, 46

No symptom

Mild elevation of urinary free cortisol

Adrenal incidentaloma

No ligand-binding activity

No transcriptional activity

(93)

G679S

2035G>A

Heterozygous

Point Mutation

F, 19

Hirsutism

Fatigue

Hypertension

Decreased affinity to ligand

Decreased transcriptional activity

(111)

R714Q

2141G>A

Heterozygous

Point Mutation

F, 2

Hypertension

Mild clitoromegaly

Advanced bone age

Precocious puberty

Hypokalemia

Decreased affinity to ligand

Decreased transcriptional activity

(84)

R714Q

2141G>A

Heterozygous

Point Mutation

F, 31

Unsuccessful attempts for pregnancy for 2.5 years

Decreased affinity to ligand

Decreased transcriptional activity

(112)

R714Q

2141G>A

Compound heterozygous with 592G>T mutation

Point Mutation

F, 3

Hypoglycemia

Hypertension

Also harbors E198X expressed from the other allele

(86)

H726R

2177A>G

Heterozygous

Point Mutation

F, 30

Hirsutism

Acne

Alopecia

Anxiety

Fatigue

Irregular menstrual cycles

Decreased affinity to ligand

Decreased transcriptional activity

(89)

V729I

2185G>A

Homozygous

Point Mutation

M, 6

Precocious puberty

Hyperandrogenism

Reduced affinity to ligand

Reduced transcriptional activity

(101)

F737L

2209T>C

Heterozygous

Point Mutation

M, 7

Hypertension

Hypokalemia

Decreased affinity to ligand

Decreased transcriptional activity

(7)

I747M

2241T>G

Heterozygous

Point Mutation

F, 18

Hirsutism

Oligo/amenorrhea

Decreased affinity to ligand

Decreased transcriptional activity

(102)

I757V

2269A>G

Heterozygous

Point Mutation

F, 23

No symptoms

Decreased affinity to ligand

Decreased transcriptional activity

(97)

L773P

2318T>C

Heterozygous

Point Mutation

F, 29

Hypertension

Hirsutism

Fatigue

Anxiety

Decreased affinity to ligand

Decreased transcriptional activity

(90)

L773VfsX25

2317-2318delCT

Heterozygous

Frame Shift

M, 27

Hypoglycemia

Fatigability with feeding

Hypertension

No ligand-binding activity

(113)

F774SfsX24

2318-2319delTG

Homozygous

Frame Shift

M, 1

Hypokalemia

Hypoglycemia

Hypertension

No ligand-binding activity

(85)

Intronic Mutations

NR (No protein expression)

1891-1894delGAGT

Heterozygous

Destruction of the splice donor site

F, 26

Hirsutism,

Menstrual Irregularities

No GR mRNA and protein expression from the affected allele

(103)

N.D.

Predicted to generate V675GfsX10

2024G > T

Heterozygous

Predicted to skip exon 8

F, 49

Hirsutism,

Menstrual Irregularities, Anxiety

N.D.

 

(91)

 

*: The case also harbors a heterozygous mutation in the 21-hydroxylase gene.

:  The 1201G>T D401H mutation causes mild glucocorticoid hypersensitivity.

N.D.; not determined。

Figure 3. Location of the NR3C1 gene mutations that cause primary generalized glucocorticoid resistance syndrome†. Currently, 36 independent mutations are reported. The mutations identified in the coding sequence of LBD, HR, DBD and NTD are shown in a light green, green, yellow and red box, respectively. Miss-sense mutations, non-sense mutations and frame-shift mutations are shown with black, purple and blue letters, respectively. Two mutations identified in the intronic sequence are shown with red letters. Homozygous mutations are shown with underlines. †: The 1201G>T D410H mutation causes mild glucocorticoid hypersensitivity; *: The same miss-sense mutation but found in unrelated subjects/families; $: Prediction only (the mutated hGR protein was not biologically identified); #: These two mutations were found as compound heterozygous in one affected subject. Numbers of nucleotides and amino acids are based on the transcription initiation site and the first methionine of the hGR protein, respectively. DBD: DNA-binding domain; HR: hinge region; LBD: ligand-binding domain; NTD: N-terminal domain

 

Molecular Defects of Pathologic hGRa Mutants

 

Molecular defects of pathologic mutant receptors have been extensively investigated by focusing on their defects in ligand-association, transactivation of glucocorticoid-responsive genes, cytoplasmic to nuclear translocation, and others (5). Recently, computer-based in silico structural simulation has also been used for estimating the structural impact of mutations to hGRa LBD and DBD (88, 114).

 

Pathologic mutant receptors generally cause inactivation/reduction of one or some of the receptor functions, whereas they are in most cases heterozygous mutations that enables affected subjects to harbor both mutated and intact hGRaprotein in their tissues (5, 6). Thus, affected subjects of this syndrome demonstrate partial loss of glucocorticoid actions in their tissues, consistent with the experimental evidence that genetic knock-out (inactivation) of the Nr3c1 gene in mice (thus, complete abbreviation of the GR protein and its actions) is lethal (115). However, one homozygous case who only expresses a mutant receptor with complete loss of the ligand-binding activity was reported (2318-2319delTG F774SfsX24) (85). Given that the ligand-binding is essential for subsequent receptor activation, this mutant receptor might have residual activities including minimal association to glucocorticoids or other steroids, enabling the patient to survive even though he only expresses this highly damaged receptor.

 

LBD MUTATIONS

 

There are 22 pathologic mutations whose amino acid changes are identified in the LBD. Among them, 17 are miss-sense mutations (see Table 1 and Figure 3 for details), one is a non-sense mutation (1992A>T Y660X) (115), and four are frame-shift mutations (1762-1765insTTAC>G H588LfsX5, 1835delC S612YfsX15, 2317-2318delCT L773VfsX25 and 2318-2319delTG F774SfsX24) (85, 92, 109, 113). Since LBD is the domain harboring a majority of receptor functions with established evaluation means (15), molecular defects of these mutant receptors have been most extensively and systemically investigated. These molecular examinations include: i) the affinity of the mutant receptors for the ligand (the synthetic pure glucocorticoid dexamethasone was used in most cases, thus the method is called “dexamethasone binding assay”); ii) the transcriptional activity of the mutant receptors on endogenous glucocorticoid-responsive genes and/or transiently introduced exogenous GRE-driven reporters; iii) the ability of in vitro physical interaction of the mutant receptors with p160-type nuclear receptor coactivators, such as the glucocorticoid receptor-interacting protein 1 (GRIP1 or NCoA2); iv) the subcellular localization of the mutant receptors and their nuclear translocation in response to glucocorticoids (in most cases, dexamethasone was used as a ligand); v) the ability of the mutant receptors to bind endogenous DNA GREs (using the chromatin-immunoprecipitation (ChIP) assay); vi) the structural analysis on the mutant receptors’ LBDs by employing the computer-based in silico three-dimensional (3D) simulation using as a template crystallographic data of the LBD peptide; vii) the motility of the mutant receptors inside the nucleus using the fluorescence recovery after photobleaching (FRAP) analysis.

 

Molecular defects in two major functions of the hGRa, the ability to bind glucocorticoids and the transactivation of glucocorticoid-responsive genes are summarized in Table 1. Compared with the wild-type receptor, all mutant receptors demonstrate variable reduction in their affinity to dexamethasone, and attenuate their transactivation of GREs-driven genes following exposure to this steroid, with the most severe impairment observed in the cases of I559N, V571A, D641V, L672P, R714Q, I747M, L773P, L773VfsX25 and F774fsX24 mutations (80, 82, 84, 85, 96, 99, 100, 102, 110, 113). In the in silico 3D structural simulation analysis on LBD of the miss-sense point mutant receptors, most of the replaced amino acids are located outside the molecular structures, which directly mediate these two major functions, LBP and the AF-2 surface, respectively (114). The latter is used for physical interaction with the LxxLL motif of p160-type coactivators (58). Further analysis revealed that these point mutations damage and/or alter multiple intramolecular amino acid interactions necessary for maintaining the proper structural conformation of LBD, resulting in the alteration in these two protein surfaces indirectly but simultaneously (114). More detailed structural analysis revealed that the amino acid replacements damage LBP by indirectly reducing the electrostatic interaction between key residues of LBP and those of the dexamethasone molecule (especially, the interaction formed against the carbonyl oxygen of carbon (C) 3 of this steroid) (114). Their impact on the interaction between the AF-2 surface and the LxxLL motif of the p160-type coactivator GRIP1 protein is variable, but tends to damage the ionic interaction (or salt bridge) of non-core leucines of this motif as well as the noncovalent interaction of its core leucine residues formed against key amino acids of the AF-2 surface, ultimately reducing the affinity of this motif to the hydrophobic cleft of the AF-2 surface (114).

 

The C-terminal portion of the hGRa LBD that follows the a-helix-12 of this domain is one of the hot spots of pathologic hGRa mutations, as evident in the accumulation of three independent mutations to this region (L773P, L773VfsX25 and F774fsX24) (85, 90, 113). Indeed, this molecular area is particularly important for creating the AF-2 surface and for maintaining the ligand-bound LPB conformation through its dramatic intramolecular shift upon binding to a ligand (30). Arginine (R) at amino acid position 714 is another hot spot of the point mutations, as three patients independently harbor this mutation that replaces this amino acid to glutamine (Q) (84, 86, 112). In the structural simulation analysis on the R714Q mutant receptor, substitution of R for Q in LBD causes a rearrangement of the side chains resulting in forming a new salt bridge between R704 and D662 and displacing Q714 (84). This relaxes some constraint on the helix-10 and results in structural changes throughout the LBD, indirectly damaging conformation of both LBP and the AF-2 surface (84). Interestingly, the third case with the R714Q mutation harbors another point mutation (592G>T E198X) in the other allele (compound heterozygous), which generates a truncated receptor at E198 (E198X) (86). Thus, the patient expresses both R714Q and E198X mutant receptors but no intact receptor in her tissues.

 

The LBD mutant receptors frequently demonstrate delay of their translocation from the cytoplasm to the nucleus compared to the wild-type receptor, consistent with the fact that the ligand-binding “turns on” the nuclear translocation of the receptor by inducing the conformational change that allows the receptor to expose NL1 and NL2 surfaces to their counterpart nuclear import systems (7, 84, 85, 89, 90, 98-100, 102, 116). Although detailed molecular mechanisms underlying this defect have not been examined yet, it is likely that the mutations interrupt proper functions of these domains (32). Some mutant receptors, such as hGRaV729I and hGRaF737L, shift their subcellular localization toward the nucleus in the absence of ligand (7, 100), possibly by their defective intracellular circulation, such as through defective NL1 activity and/or altered interaction with14-3-3 proteins, calreticulin or others.

 

All LBD mutant receptors tested for their interaction with DNA GREs preserve their ability to bind this recognition sequences, because they have intact DBD, which can function independently to LBD (7, 84, 85, 89, 90, 98-100, 102, 116). Further, many of these mutant receptors demonstrate a dominant negative effect on the transcriptional activity of the wild-type receptor, because they are in most cases partially active mutants, and thus, can interfere with the full activity of the wild-type receptor, such as by competing for the molecules mediating the latter’s transcriptional activity (e.g., by squelching transcriptional cofactors including p160-type coactivators) (5, 6, 102). Finally, the LBD point mutant receptors tested in the FRAP analysis demonstrate dynamic motility defects inside the nucleus of living cells, possibly due to their reduced affinity to ligand and/or inability to interact properly with key cofactors and/or chromatin molecules (117).

 

Molecular characterization of the LBD mutants explained above have been performed mostly by employing cell-based bioassays. However, Kaziales, et. al., recently performed in vitro biochemical assays on the L773P mutant receptor by employing its purified peptide consisting of DBD, HR, and intact or mutated LBD (118). The “wild-type” receptor peptide (called GRm) employed for their assays harbors multiple amino acid replacements for conferring its peptide stability. Thus, the authors compared GRm and GRmL773P, and found that the latter has altered physical interaction with HSP90 (118). They suggested that this molecular defect underlies the reduced interaction of the receptor peptide to dexamethasone, the LxxLL motif, and further, DNA GREs, although exact molecular evidence and associated mechanisms were not demonstrated.

 

HR MUTATIONS

 

Two pathologic mutations were identified in HR (Table 1 and Figure 3). One is a non-sense mutation (1471 C>T R491X) and the other is a miss-sense mutation (1503 G>T Q501H) (97). Both are located in exon 5. The patient harboring R491X developed typical manifestations of Chrousos syndrome, as the mutant receptor lacks the entire LBD (97). On the other hand, the subject harboring Q501H demonstrated biochemical changes only, while the mutant receptor showed weakly reduced transactivation of the exogenous glucocorticoid-responsive gene (97).

 

DBD MUTATIONS

 

Currently, seven pathologic mutations were identified in DBD (Table 1 and Figure 3). Among them, five are miss-sense (point) mutations. The other two are a non-sense mutation and a frame-shift mutation. All five point mutant receptors reduce or lose their affinity to DNA GREs  (88, 92, 93, 97, 107). In contrast, they retain intact affinity for ligand dexamethasone, because DBD and LBD function independently with each other (88, 92, 93, 97, 107). Among these point mutations, three (1430G>A R477H, 1429C>T R477S and 1429C>T R477C) replace arginine (R) at amino acid position 477 to other amino acids (histidine (H), serine (S) and cysteine (C), respectively), while one targets tyrosine (Y) at position 478 and changes it to cysteine (C) (1433A>G Y478C). Thus, the area around R477 and Y478 appears to be a hot-spot of DBD mutations. These two amino acids are located just C-terminally to the fourth cysteine residue of the second zinc finger of DBD, which participates in holding a zinc ion together with the other three cysteines of this finger motif. R477 is critical for maintaining the ability of the receptor to bind GREs by providing the hydrophobicity required for its interaction with the backbone chain of the GRE DNA. Thus, replacement of either of these two amino acids seems to reduce the affinity of the mutant receptors to the GRE DNA through damaging this local hydrophobicity.

 

The point mutation 1268T>C V423A replaces valine (V) at amino acid position 423 to alanine (A) (88). V423 is located just N-terminally to the second cysteine of the first zinc finger of DBD. Replacement of this valine to alanine at amino acid position 423 permits water molecules to diffuse into the zinc-binding region of the receptor and indirectly damages the hydrophobicity maintained by R477, leading to the reduction in the affinity of this mutant receptor to the GRE DNA (88).

 

Interestingly, the mutant receptors V423A, R477S and Y478C demonstrate delayed cytoplasmic to nuclear translocation upon exposure to dexamethasone (88, 93). Molecular defect(s) underlying this impairment have(s) not been elucidated, but these mutations appear to affect indirectly the function of NL1, because this molecular surface spans over the second zinc finger of DBD, while these mutations damage the hydrophobic circumstance around this finger (88, 107). The second zinc finger of the DBD is also critical for receptor homodimerization, which is a prerequisite for the receptor to bind a tandem GREs and subsequent transactivation of glucocorticoid-responsive genes harboring this DNA sequence (119). Thus, defective homodimerization may also contribute to the reduced transcriptional activity of these DBD mutant receptors.

 

NTD MUTATIONS

 

Four independent point mutations are reported in NTD. These include 26C>G P9R, 367G>T Q123X, 592G>T E198X and 1201G>T D401H (86, 87, 104, 105). The 367G>T Q123X and the 592G>T E198X are non-sense mutations generating truncated receptors, respectively at amino acid position 123 and 198. Because both receptors appear to be highly damaged as they lack the entire DBD and LBD, the affected subjects demonstrated clear-cut manifestations of Chrousos syndrome (86, 87).

 

The patient harboring the 26C>G P9R mutation demonstrated mild clinical manifestations with slight increase in ACTH and cortisol secretion (104). Molecular characterization was not performed for this mutant receptor (104), thus there is a possibility that the identified nucleotide change is not pathologic. Indeed, NTD (exon 2) is the domain most harboring single nucleotide polymorphisms (SNPs) among all three major domains throughout the nuclear hormone receptor genes (13), thus this domain can well tolerate to nucleotide replacements and tends to maintain its proper functions compared to the other domains.

 

The patient harboring the 1201G>T D401H mutation demonstrated mild hypersensitivity to glucocorticoids in contrast to the other pathologic mutations that cause glucocorticoid resistance (105). Compared to the wild-type receptor, the D401H mutant receptor demonstrated ~2-fold stronger transcriptional activity in a reporter assay, which is equivalent to the activity of the N363S mutant receptor in a side-by-side assay. The nucleotide change causing the N363S replacement is a well-known polymorphism associated with mild glucocorticoid hypersensitivity (120-122). Thus, the 1201G>T D401H may be another weakly functional polymorphism causing mild tissue hypersensitivity to glucocorticoids.

 

The 3-year-old girl with the 592G>T E198X mutation additionally harbors the 2141G>A R714Q mutation in the other allele as explained in the section of “LBD mutations” (86). She developed severe manifestations of glucocorticoid resistance, such as uncontrollable hypertension, brain micro-infarctions, and hypoglycemic coma, because both mutant receptors she harbored are highly damaged. The family study revealed that the 592G>T E198X mutation is maintained among her family, while the 2141G>A R714Q mutation is de novo in the affected girl (86).

 

INTRONIC MUTATIONS

 

So far, only two intronic mutations are reported. One is the 1891-1894 delGAGT NR, which deletes four nucleotides (GAGT) at the nucleotide position 1891-1894 (in intron F located between exon 4 and 5) that destroys the intron-acceptor site located 5’ terminally to exon 6 (91, 103). The mutated mRNA expressed from the affected allele loses its biological stability, therefore the mutation functionally “knocks-out” NR3C1 of this allele (103). The amount of patient’s tissue hGRa is thus 50% of the healthy subjects as the receptor protein is only expressed from the intact allele (103). The other mutation is the 2024G>T, which replaces G with T at the position one nucleotide 5’ terminally to exon 8 (thus, located at the 3’-terminal portion of intron I) (91). Although biochemical characterization on the mutant receptor was not performed, the computer-based prediction indicated that the mutation appears to cause a skip of the entire exon 8 and to generate the V675GfsX10 truncated receptor whose molecular function appears to be highly damaged (91). It is also possible that the mutation reduces stability of its mRNA, leading to functional “knock-out” of NR3C1 of the affected allele similar to the 1891-1894 delGAGT NR mutation (103). Thus, biochemical evaluation on the mutated mRNA and hGRaprotein is needed.

             

Clinical Evaluation of Patients with Primary Generalized Glucocorticoid Resistance Syndrome

 

Key for evaluating patients with this syndrome is to identify the manifestations suggesting upregulation of the HPA axis without Cushingoid features (5) (Table 2). Circadian rhythmicity of circulating ACTH and cortisol should be preserved, in contrast to the patients with Cushing syndrome (5). In addition, any evidence suggesting psychiatric problems (e.g., anxiety and depression), possibly through upregulation of brain CRH and/or AVP may be noted (5).

 

Physical examination should include an assessment for signs of hypertension and associated metabolic alkalosis caused by elevated levels of adrenal mineralocorticoids (5). Arterial blood pressure should be recorded and should be monitored over a 24-hour period. Signs of hyperandrogenism and/or virilization caused by over-production of the adrenal androgens, such as acne, hirsutism, pubic and axillary hair development, male-pattern hair loss, and clitoromegaly, should be evaluated. Hirsutism should be assessed using the Ferriman-Gallwey score (123), while pubic hair development should be classified according to the Tanner scale (124, 125). All subjects should be screened for signs associated with Cushing syndrome or therapeutic use of high-dose glucocorticoids.

 

Table 2. Clinical Manifestations and Diagnostic Evaluation of Primary Generalized Glucocorticoid Resistance Syndrome

Clinical Presentation

Glucocorticoid excess

Apparently normal glucocorticoid actions without Cushingoid features

(However, hypoglycemia and associated coma/seizures can be observed in affected neonates/young children)

 

Mineralocorticoid excess

                   Hypertension

                   Hypokalemic alkalosis

 

Adrenal androgen excess

Children: Ambiguous genitalia at birth*, clitoromegaly, premature adrenarche, gonadotropin-independent precocious puberty

Females: Acne, hirsutism, male-pattern hair loss, menstrual irregularities, oligo-anovulation, infertility

Males: Acne, hirsutism, oligospermia, adrenal rests in the testes, infertility

 

CRH/AVP excess in brain hypothalamus and elevation of circulating ACTH levels

Anxiety

Benign pituitary tumors (ACTH-producing)

Bilateral adrenal hyperplasia

Adrenal adenomas

 

Diagnostic Evaluation

Upward shift of the HPA axis activity and responsiveness to high-dose glucocorticoids

Elevated plasma ACTH concentrations

Elevated serum cortisol concentrations

Increased 24-hour urinary free cortisol (UFC) excretion

Resistance of the HPA axis to dexamethasone suppression but positive response to its high, grading doses

 

Normal circadian rhythmicity of circulating cortisol and ACTH concentrations

 

Presence of glucocorticoid resistance in patients’ tissues

The thymidine incorporation assay using patients’ PBMCs: Reduced dexamethasone-induced suppression of phytohemagglutinin-stimulated thymidine incorporation compared to normal subjects

The dexamethasone binding assay using patients’ PBMCs: Decreased affinity to dexamethasone compared to normal subjects

 

Identification of mutation(s) in the NR3C1 gene (mostly in its coding sequence but can be in exon/intron junctions)

 

Identification of molecular defects of mutant receptors in appropriate assay systems

 

* The case demonstrating this manifestation also harbored a heterozygous mutation in the 21-hydroxylase gene.

 

Endocrinological Evaluation of Patients with Primary Generalized Glucocorticoid Resistance Syndrome

 

The aim of the endocrinological evaluation is to demonstrate up-regulation of the HPA axis with preservation of its normal circadian rhythmicity and blunted responsiveness to exogenous glucocorticoids (5). Concentrations of plasma ACTH, renin activity and aldosterone, as well as serum cortisol, corticosterone, deoxycorticosterone, testosterone, androstenedione, DHEA, and DHEA-S should be measured. Determination of 24-hour UFC excretion on 2 or 3 consecutive days is important to access the presence of hypercortisolism. Diurnal fluctuation of plasma ACTH and serum cortisol should be evaluated, for example, by monitoring them both in the morning and in the evening.

 

Responsiveness of the HPA axis to exogenous glucocorticoids should be examined using the dexamethasone suppression test (5). Increasing doses of dexamethasone (e.g., 0.3, 0.6, 1.0, 1.5, 2.0, 2.5, and 3.0 mg) should be given orally at midnight every other day, and a serum sample should be drawn at 0800h the following morning for determining serum cortisol concentrations. Affected subjects demonstrate resistance of the HPA axis to administered dexamethasone but can respond to higher doses. Concurrent measurement of serum dexamethasone concentrations is recommended in order to exclude the possibility of increased metabolic clearance or decreased absorption of this compound (83). Pituitary and adrenal imaging studies should be performed, because patients with this syndrome frequently harbor hypertrophy of these organs or may develop their benign tumors. 

 

Cellular and Molecular Studies on Patients with Primary Generalized Glucocorticoid Resistance Syndrome

 

The purpose of cellular studies is to identify the presence of tissue resistance to glucocorticoids in actual tissues of the affected subjects. The thymidine incorporation assay and the dexamethasone binding assay employing subjects’ peripheral blood mononuclear cells (PBMCs) are generally employed (5, 126) (Table 2). In the former assay, dexamethasone administration strongly suppresses phytohemagglutinin-stimulated thymidine incorporation of PBMCs in normal subjects. However, this response is significantly blunted in the affected subjects due to reduced affinity/actions of this steroid in these cells. The dexamethasone binding assay can address reduction in the affinity of patients’ tissue hGRa to dexamethasone, because mutant receptors harboring their defects in LBD almost always show reduced affinity for this steroid.

 

As part of the molecular examination for verifying pathologic causes and their molecular mechanisms, sequencing of the coding region of the NR3C1 gene including exon/intron junctions should be performed (126). Identification of mutations in the NR3C1 gene is critical for diagnosing this syndrome. Once mutations are identified, the next step is to prove that the identified mutations have biologic impact. Because the NR3C1 gene harbors so many neutral polymorphisms (13), there is always a possibility that the identified nucleotide changes are just coincidental but not pathologic. Population incidence of the identified nucleotide changes is important if available, as pathologic mutations generally have a very low allele frequency. Molecular studies can be started by constructing the mutant hGRa-expressing plasmids. Then, molecular actions of mutant receptors can be examined by transfecting the created plasmids (transiently or stably) to appropriate cell lines (e.g., GR-negative African green monkey kidney CV1 and COS7 cells, and GR-positive human cervical cancer HeLa cells). Using mutant receptor-expressing cultured cells, reporter transactivation assays using the GREs-driven luciferase gene can be performed to address the reduced transcriptional activity of mutant receptors. The dexamethasone binding assay can also be performed in the COS7 cells transiently expressing mutant receptors to evaluate their affinity to dexamethasone in the absence of the wild-type GR. In microscope-based imaging studies on the cells transfected with plasmids expressing mutant receptors, their abnormal subcellular localization and delayed nuclear translocation in response to dexamethasone can be evaluated.

 

Management of Patients with Primary Generalized Glucocorticoid Resistance Syndrome

 

The aim of the treatment for patients with this syndrome is to suppress the excess ACTH secretion in order to reduce production of the adrenal steroids with mineralocorticoid and/or androgenic activity to minimize their pathologic effects (5). Treatment involves the administration of high doses of mineralocorticoid activity-sparing pure glucocorticoids (e.g., dexamethasone), which activate mutated and/or wild-type hGRα in the hypothalamus/pituitary gland of the affected subjects and suppress their ACTH secretion. Adequate suppression of the HPA axis is of particular importance, given that the treatment is virtually life-long, thus any side effects of exogenous glucocorticoids should be avoided as much as possible. Long-term dexamethasone treatment should be titrated carefully according to the clinical manifestations and biochemical profiles of the affected subjects.

 

CONCLUSIVE REMARKS AND FUTURE PERSPECTIVES

 

Primary generalized glucocorticoid resistance syndrome is characterized by hypercortisolism without Cushingoid features but with manifestations caused by upregulation of the HPA axis, such as hypertension (by mineralocorticoid excess) and signs of hyperandrogenism (by adrenal androgen excess) (81). The pathologic cause of this syndrome is ascribed to mutations in the NR3C1 gene, which decrease the action of its encoding protein hGRa, a ligand-dependent transcription factor (15, 81). In honor to Professor George P. Chrousos who discovered the first case and significantly contributed to the progress of this field, this syndrome may be called “Chrousos syndrome”, particularly for the cases who demonstrate classic and characteristic manifestations of this syndrome (6, 80). Recent progress in genome technology including high through-put sequencing has enabled clinical researchers to handle large patient cohorts and clinicians can get access to the NR3C1 gene sequencing much easier and faster than before. Consequently, 35 cases/families of this syndrome are currently reported world-wide who harbor pathologic mutations in the NR3C1 gene. It is of note that some of the recent cases tend to demonstrate much milder manifestations compared to the classic cases of Chrousos syndrome (97, 110). Further, some of them even lack obvious manifestations but show biochemical or imaging abnormalities only (93, 97). For these cases with very mild or no manifestations, their genetic changes may be considered as rare polymorphisms rather than pathologic mutations. Further discussion is needed for distinguishing pathologic mutations and mildly functional polymorphisms based on their clinical manifestations and allele frequency of the nucleotide changes. 

 

In some reported cases, molecular defects of the mutated receptors were not evaluated. Testing them in tandem with the wild-type receptor is crucial for avoiding false-diagnosis, because the NR3C1 gene harbor substantial numbers of biologically silent polymorphisms (13). On the other hand, there are patients who demonstrate characteristic manifestations of Chrousos syndrome but do not harbor pathologic mutations in the NR3C1 gene. These “mutation-silent” subjects might carry their genetic defects not in NR3C1 but in other genes whose encoding proteins function in the glucocorticoid signaling pathway. For example, there was a boy who demonstrated manifestations compatible with multiple steroid hormone resistance (127). He harbored a small gene segmental deletion around one zinc finger protein (ZNF) gene, and Its encoding protein ZNF764 turned out to function as a coactivator of several steroid hormone receptors including the hGRa (127). As our knowledge of the glucocorticoid signaling pathway increases, including new players like long non-coding RNAs (15, 128, 129), we hope that genetic cause(s) of undiagnosed cases with Chrousos syndrome will soon be identified, by employing classic genetic methods (e.g., the linkage analysis) as well as cutting-edge genome-related methodologies including the whole genome/exome sequencing and sophisticated bioinformatical/statistical analysis tools.

 

REFERENCES

 

  1. Chrousos GP. Stress and disorders of the stress system. Nat Rev Endocrinology. 2009;5(7):374-381.
  2. Chrousos GP, Charmandari E, and Kino T. Glucocorticoid action networks -an introduction to systems biology. J Clin Endocrinol Metab. 2004;89(2):563-564.
  3. Kino T, De Martino MU, Charmandari E, Mirani M, and Chrousos GP. Tissue glucocorticoid resistance/hypersensitivity syndromes. J Steroid Biochem Mol Biol. 2003;85(2-5):457-467.
  4. Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med. 1995;332(20):1351-1362.
  5. Charmandari E, Kino T, Ichijo T, and Chrousos GP. Generalized glucocorticoid resistance: clinical aspects, molecular mechanisms, and implications of a rare genetic disorder. J Clin Endocrinol Metab. 2008;93(5):1563-1572.
  6. Charmandari E, and Kino T. Chrousos syndrome: a seminal report, a phylogenetic enigma and the clinical implications of glucocorticoid signalling changes. Eur J Clin Invest. 2010;40(10):932-942.
  7. Charmandari E, Kino T, Ichijo T, Jubiz W, Mejia L, Zachman K, et al. A novel point mutation in helix 11 of the ligand-binding domain of the human glucocorticoid receptor gene causing generalized glucocorticoid resistance. J Clin Endocrinol Metab. 2007;92(10):3986-3990.
  8. Kino T, and Chrousos GP. In: Steckler T, Kalin NH, and Reul JMHM eds. Handbook on Stress and the Brain. Amsterdam: Elsevier BV; 2005:295-312.
  9. Chrousos GP. The glucocorticoid receptor gene, longevity, and the complex disorders of Western societies. Am J Med. 2004;117(3):204-207.
  10. Nicolaides NC, Galata Z, Kino T, Chrousos GP, and Charmandari E. The human glucocorticoid receptor: molecular basis of biologic function. Steroids. 2010;75(1):1-12.
  11. Nuclear Receptors Nomenclature Committee. A unified nomenclature system for the nuclear receptor superfamily. Cell. 1999;97(2):161-163.
  12. Germain P, Staels B, Dacquet C, Spedding M, and Laudet V. Overview of nomenclature of nuclear receptors. Pharmacol Rev. 2006;58(4):685-704.
  13. Mackeh R, Marr AK, Dargham SR, Syed N, Fakhro KA, and Kino T. Single-nucleotide variations of the human nuclear hormone receptor genes in 60,000 individuals. J Endocr Soc. 2018;2(1):77-90.
  14. Chrousos GP, and Kino T. Intracellular glucocorticoid signaling: a formerly simple system turns stochastic. Sci STKE. 2005;2005(304):pe48.
  15. Nicolaides NC, Chrousos G, and Kino T. Glucocorticoid Receptor. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al. eds. Endotext. South Dartmouth (MA): MDText.com, Inc. Copyright © 2000-2024, MDText.com, Inc.; 2000.
  16. Bamberger CM, Bamberger AM, de Castro M, and Chrousos GP. Glucocorticoid receptor b, a potential endogenous inhibitor of glucocorticoid action in humans. J Clin Invest. 1995;95(6):2435-2441.
  17. Lewis-Tuffin LJ, and Cidlowski JA. The physiology of human glucocorticoid receptor b (hGRb) and glucocorticoid resistance. Ann N Y Acad Sci. 2006;1069:1-9.
  18. Oakley RH, Jewell CM, Yudt MR, Bofetiado DM, and Cidlowski JA. The dominant negative activity of the human glucocorticoid receptor b isoform. Specificity and mechanisms of action. J Biol Chem. 1999;274(39):27857-27866.
  19. Kino T, Su YA, and Chrousos GP. Human glucocorticoid receptor isoform b: Recent understanding of its potential implications in physiology and pathophysiology. Cell Mol Life Sci. 2009;66(21):3435-3448.
  20. Ramos-Ramírez P, and Tliba O. Glucocorticoid receptor β (GRβ): Beyond its dominant-negative function. Int J Mol Sci. 2021;22(7);3649.
  21. Stechschulte LA, Wuescher L, Marino JS, Hill JW, Eng C, and Hinds TD, Jr. Glucocorticoid receptor β stimulates Akt1 growth pathway by attenuation of PTEN. J Biol Chem. 2014;289(25):17885-17894.
  22. Yin Y, Zhang X, Li Z, Deng L, Jiao G, Zhang B, et al. Glucocorticoid receptor β regulates injury-mediated astrocyte activation and contributes to glioma pathogenesis via modulation of β-catenin/TCF transcriptional activity. Neurobiol Dis. 2013;59:165-176.
  23. Wang Q, Lu PH, Shi ZF, Xu YJ, Xiang J, Wang YX, et al. Glucocorticoid receptor β acts as a co-activator of T-cell factor 4 and enhances glioma cell proliferation. Mol Neurobiol. 2015;52(3):1106-1118.
  24. Lu NZ, and Cidlowski JA. Translational regulatory mechanisms generate N-terminal glucocorticoid receptor isoforms with unique transcriptional target genes. Mol Cell. 2005;18(3):331-342.
  25. Presul E, Schmidt S, Kofler R, and Helmberg A. Identification, tissue expression, and glucocorticoid responsiveness of alternative first exons of the human glucocorticoid receptor. J Mol Endocrinol. 2007;38(1-2):79-90.
  26. Turner JD, and Muller CP. Structure of the glucocorticoid receptor (NR3C1) gene 5' untranslated region: Identification, and tissue distribution of multiple new human exon 1. J Mol Endocrinol. 2005;35(2):283-292.
  27. Sinclair D, Fullerton JM, Webster MJ, and Shannon Weickert C. Glucocorticoid receptor 1B and 1C mRNA transcript alterations in schizophrenia and bipolar disorder, and their possible regulation by GR gene variants. PLoS One. 2012;7(3):e31720.
  28. Sinclair D, Webster MJ, Fullerton JM, and Weickert CS. Glucocorticoid receptor mRNA and protein isoform alterations in the orbitofrontal cortex in schizophrenia and bipolar disorder. BMC Psychiatry. 2012;12:84.
  29. Frank F, Ortlund EA, and Liu X. Structural insights into glucocorticoid receptor function. Biochem Soc Trans. 2021;49(5):2333-2343.
  30. Bledsoe RK, Montana VG, Stanley TB, Delves CJ, Apolito CJ, McKee DD, et al. Crystal structure of the glucocorticoid receptor ligand binding domain reveals a novel mode of receptor dimerization and coactivator recognition. Cell. 2002;110(1):93-105.
  31. Pratt WB. The role of heat shock proteins in regulating the function, folding, and trafficking of the glucocorticoid receptor. J Biol Chem. 1993;268(29):21455-21458.
  32. Savory JG, Hsu B, Laquian IR, Giffin W, Reich T, Haché RJ, et al. Discrimination between NL1- and NL2-mediated nuclear localization of the glucocorticoid receptor. Mol Cell Biol. 1999;19(2):1025-1037.
  33. Hoelz A, Debler EW, and Blobel G. The structure of the nuclear pore complex. Annu Rev Biochem. 2011;80:613-643.
  34. Terry LJ, Shows EB, and Wente SR. Crossing the nuclear envelope: hierarchical regulation of nucleocytoplasmic transport. Science. 2007;318(5855):1412-1416.
  35. Kinyamu HK, Chen J, and Archer TK. Linking the ubiquitin-proteasome pathway to chromatin remodeling/modification by nuclear receptors. J Mol Endocrinol. 2005;34(2):281-297.
  36. Kino T, Tiulpakov A, Ichijo T, Chheng L, Kozasa T, and Chrousos GP. G protein b interacts with the glucocorticoid receptor and suppresses its transcriptional activity in the nucleus. J Cell Biol. 2005;169(6):885-896.
  37. Kino T, Souvatzoglou E, Charmandari E, Ichijo T, Driggers P, Mayers C, et al. Rho family guanine nucleotide exchange factor Brx couples extracellular signals to the glucocorticoid signaling system. J Biol Chem. 2006;281(14):9118-9126.
  38. Boldizsar F, Szabo M, Kvell K, Czompoly T, Talaber G, Bjorkan J, et al. ZAP-70 tyrosines 315 and 492 transmit non-genomic glucocorticoid (GC) effects in T cells. Mol Immunol. 2013;53(1-2):111-117.
  39. Kokkinopoulou I, and Moutsatsou P. Mitochondrial glucocorticoid receptors and their actions. Int J Mol Sci. 2021;22(11):6054.
  40. Haché RJ, Tse R, Reich T, Savory JG, and Lefebvre YA. Nucleocytoplasmic trafficking of steroid-free glucocorticoid receptor. J Biol Chem. 1999;274(3):1432-1439.
  41. Black BE, Holaska JM, Rastinejad F, and Paschal BM. DNA binding domains in diverse nuclear receptors function as nuclear export signals. Curr Biol. 2001;11(22):1749-1758.
  42. Holaska JM, Black BE, Love DC, Hanover JA, Leszyk J, and Paschal BM. Calreticulin is a receptor for nuclear export. J Cell Biol. 2001;152(1):127-140.
  43. Holaska JM, Black BE, Rastinejad F, and Paschal BM. Ca2+-dependent nuclear export mediated by calreticulin. Mol Cell Biol. 2002;22(17):6286-6297.
  44. Kino T, Souvatzoglou E, De Martino MU, Tsopanomihalu M, Wan Y, and Chrousos GP. Protein 14-3-3s interacts with and favors cytoplasmic subcellular localization of the glucocorticoid receptor, acting as a negative regulator of the glucocorticoid signaling pathway. J Biol Chem. 2003;278(28):25651-25656.
  45. Habib T, Sadoun A, Nader N, Suzuki S, Liu W, Jithesh PV, et al. AKT1 has dual actions on the glucocorticoid receptor by cooperating with 14-3-3. Mol Cell Endocrinol. 2017;439:431-443.
  46. Kino T. GR-regulating serine/threonine kinases: New physiologic and pathologic implications. Trends Endocrinol Metab. 2018;29(4):260-270.
  47. Piovan E, Yu J, Tosello V, Herranz D, Ambesi-Impiombato A, Da Silva AC, et al. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell. 2013;24(6):766-776.
  48. Munier CC, De Maria L, Edman K, Gunnarsson A, Longo M, MacKintosh C, et al. Glucocorticoid receptor Thr524 phosphorylation by MINK1 induces interactions with 14-3-3 protein regulators. J Biol Chem. 2021;296:100551.
  49. Ramamoorthy S, and Cidlowski JA. Exploring the molecular mechanisms of glucocorticoid receptor action from sensitivity to resistance. Endocr Dev. 2013;24:41-56.
  50. Schaaf MJ, and Cidlowski JA. Molecular mechanisms of glucocorticoid action and resistance. J Steroid Biochem Mol Biol. 2002;83(1-5):37-48.
  51. Beato M, and Sánchez-Pacheco A. Interaction of steroid hormone receptors with the transcription initiation complex. Endocr Rev. 1996;17(6):587-609.
  52. McKenna NJ, and O'Malley BW. Combinatorial control of gene expression by nuclear receptors and coregulators. Cell. 2002;108(4):465-474.
  53. Osz J, Brélivet Y, Peluso-Iltis C, Cura V, Eiler S, Ruff M, et al. Structural basis for a molecular allosteric control mechanism of cofactor binding to nuclear receptors. Proc Natl Acad Sci U S A. 2012;109(10):E588-594.
  54. Bulynko YA, and O'Malley BW. Nuclear receptor coactivators: Structural and functional biochemistry. Biochemistry. 2011;50(3):313-328.
  55. Mahajan MA, and Samuels HH. Nuclear hormone receptor coregulator: Role in hormone action, metabolism, growth, and development. Endocr Rev. 2005;26(4):583-597.
  56. York B, and O'Malley BW. Steroid receptor coactivator (SRC) family: Masters of systems biology. J Biol Chem. 2010;285(50):38743-38750.
  57. Heery DM, Kalkhoven E, Hoare S, and Parker MG. A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature. 1997;387(6634):733-736.
  58. Darimont BD, Wagner RL, Apriletti JW, Stallcup MR, Kushner PJ, Baxter JD, et al. Structure and specificity of nuclear receptor-coactivator interactions. Genes Dev. 1998;12(21):3343-3356.
  59. Chinenov Y, Gupte R, Dobrovolna J, Flammer JR, Liu B, Michelassi FE, et al. Role of transcriptional coregulator GRIP1 in the anti-inflammatory actions of glucocorticoids. Proc Natl Acad Sci U S A. 2012;109(29):11776-11781.
  60. Sheppard KA, Rose DW, Haque ZK, Kurokawa R, McInerney E, Westin S, et al. Transcriptional activation by NF-kB requires multiple coactivators. Mol Cell Biol. 1999;19(9):6367-6378.
  61. Richter WF, Nayak S, Iwasa J, and Taatjes DJ. The Mediator complex as a master regulator of transcription by RNA polymerase II. Nat Rev Mol Cell Biol. 2022;23(11):732-749.
  62. Colley SM, and Leedman PJ. SRA and its binding partners: an expanding role for RNA-binding coregulators in nuclear receptor-mediated gene regulation. Crit Rev Biochem Mol Biol. 2009;44(1):25-33.
  63. Colley SM, and Leedman PJ. Steroid Receptor RNA Activator - A nuclear receptor coregulator with multiple partners: Insights and challenges. Biochimie. 2011;93(11):1966-1972.
  64. Kino T, Hurt DE, Ichijo T, Nader N, and Chrousos GP. Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 2010;3(107):ra8.
  65. Gao Y, Liu C, Wu T, Liu R, Mao W, Gan X, et al. Current status and perspectives of non-coding RNA and phase separation interactions. Biosci Trends. 2022;16(5):330-345.
  66. Johnson TA, Chereji RV, Stavreva DA, Morris SA, Hager GL, and Clark DJ. Conventional and pioneer modes of glucocorticoid receptor interaction with enhancer chromatin in vivo. Nucleic Acids Res. 2018;46(1):203-214.
  67. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, et al. DNA binding of the glucocorticoid receptor is not essential for survival. Cell. 1998;93(4):531-541.
  68. Reichardt HM, Tuckermann JP, Göttlicher M, Vujic M, Weih F, Angel P, et al. Repression of inflammatory responses in the absence of DNA binding by the glucocorticoid receptor. EMBO J. 2001;20(24):7168-7173.
  69. Cruz-Topete D, and Cidlowski JA. One hormone, two actions: anti- and pro-inflammatory effects of glucocorticoids. Neuroimmunomodulation. 2015;22(1-2):20-32.
  70. Oakley RH, and Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132(5):1033-1044.
  71. Hinz B, and Hirschelmann R. Rapid non-genomic feedback effects of glucocorticoids on CRF-induced ACTH secretion in rats. Pharm Res. 2000;17(10):1273-1277.
  72. Karst H, Berger S, Turiault M, Tronche F, Schütz G, and Joëls M. Mineralocorticoid receptors are indispensable for nongenomic modulation of hippocampal glutamate transmission by corticosterone. Proc Natl Acad Sci U S A. 2005;102(52):19204-19207.
  73. Hafezi-Moghadam A, Simoncini T, Yang Z, Limbourg FP, Plumier JC, Rebsamen MC, et al. Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase. Nat Med. 2002;8(5):473-479.
  74. Löwenberg M, Verhaar AP, Bilderbeek J, Marle J, Buttgereit F, Peppelenbosch MP, et al. Glucocorticoids cause rapid dissociation of a T-cell-receptor-associated protein complex containing LCK and FYN. EMBO Rep. 2006;7(10):1023-1029.
  75. Ayroldi E, Cannarile L, Migliorati G, Nocentini G, Delfino DV, and Riccardi C. Mechanisms of the anti-inflammatory effects of glucocorticoids: Genomic and nongenomic interference with MAPK signaling pathways. FASEB J. 2012;26(12):4805-4820.
  76. Demonacos C, Djordjevic-Markovic R, Tsawdaroglou N, and Sekeris CE. The mitochondrion as a primary site of action of glucocorticoids: the interaction of the glucocorticoid receptor with mitochondrial DNA sequences showing partial similarity to the nuclear glucocorticoid responsive elements. J Steroid Biochem Mol Biol. 1995;55(1):43-55.
  77. Demonacos C, Tsawdaroglou NC, Djordjevic-Markovic R, Papalopoulou M, Galanopoulos V, Papadogeorgaki S, et al. Import of the glucocorticoid receptor into rat liver mitochondria in vivo and in vitro. J Steroid Biochem Mol Biol. 1993;46(3):401-413.
  78. Psarra AM, and Sekeris CE. Glucocorticoids induce mitochondrial gene transcription in HepG2 cells: role of the mitochondrial glucocorticoid receptor. Biochim Biophys Acta. 2011;1813(10):1814-1821.
  79. Lee SR, Kim HK, Song IS, Youm J, Dizon LA, Jeong SH, et al. Glucocorticoids and their receptors: insights into specific roles in mitochondria. Prog Biophys Mol Biol. 2013;112(1-2):44-54.
  80. Chrousos GP, Vingerhoeds A, Brandon D, Eil C, Pugeat M, DeVroede M, et al. Primary cortisol resistance in man. A glucocorticoid receptor-mediated disease. J Clin Invest. 1982;69(6):1261-1269.
  81. Chrousos G. Q&A: primary generalized glucocorticoid resistance. BMC Med. 2011;9:27.
  82. Mendonca BB, Leite MV, de Castro M, Kino T, Elias LL, Bachega TA, et al. Female pseudohermaphroditism caused by a novel homozygous missense mutation of the GR gene. J Clin Endocrinol Metab. 2002;87(4):1805-1809.
  83. Nicolaides NC, and Charmandari E. Chrousos syndrome: from molecular pathogenesis to therapeutic management. Eur J Clin Invest. 2015;45(5):504-514.
  84. Nader N, Bachrach BE, Hurt DE, Gajula S, Pittman A, Lescher R, et al. A novel point mutation in helix 10 of the human glucocorticoid receptor causes generalized glucocorticoid resistance by disrupting the structure of the ligand-binding domain. J Clin Endocrinol Metab. 2010;95(5):2281-2285.
  85. McMahon SK, Pretorius CJ, Ungerer JP, Salmon NJ, Conwell LS, Pearen MA, et al. Neonatal complete generalized glucocorticoid resistance and growth hormone deficiency caused by a novel homozygous mutation in Helix 12 of the ligand binding domain of the glucocorticoid receptor gene (NR3C1). J Clin Endocrinol Metab. 2010;95(1):297-302.
  86. Tatsi C, Xekouki P, Nioti O, Bachrach B, Belyavskaya E, Lyssikatos C, et al. A novel mutation in the glucocorticoid receptor gene as a cause of severe glucocorticoid resistance complicated by hypertensive encephalopathy. J Hypertens. 2019;37(7):1475-1481.
  87. Paragliola RM, Costella A, Corsello A, Urbani A, and Concolino P. A novel pathogenic variant in the N-terminal domain of the glucocorticoid receptor, causing glucocorticoid resistance. Mol Diagn Ther. 2020;24(4):473-485.
  88. Roberts ML, Kino T, Nicolaides NC, Hurt DE, Katsantoni E, Sertedaki A, et al. A novel point mutation in the DNA-binding domain (DBD) of the human glucocorticoid receptor causes primary generalized glucocorticoid resistance by disrupting the hydrophobic structure of its DBD. J Clin Endocrinol Metab. 2013;98(4):E790-795.
  89. Nicolaides NC, Geer EB, Vlachakis D, Roberts ML, Psarra AM, Moutsatsou P, et al. A novel mutation of the hGR gene causing Chrousos syndrome. Eur J Clin Invest. 2015;45(8):782-791.
  90. Charmandari E, Raji A, Kino T, Ichijo T, Tiulpakov A, Zachman K, et al. A novel point mutation in the ligand-binding domain (LBD) of the human glucocorticoid receptor (hGR) causing generalized glucocorticoid resistance: the importance of the C terminus of hGR LBD in conferring transactivational activity. J Clin Endocrinol Metab. 2005;90(6):3696-3705.
  91. Mauri S, Nieto-Moragas J, Obón M, and Oriola J. The glucocorticoid resistance syndrome. Two cases of a novel pathogenic variant in the glucocorticoid receptor gene. JCEM Case Rep. 2024;2(1):luad153.
  92. Velayos T, Grau G, Rica I, Pérez-Nanclares G, and Gaztambide S. Glucocorticoid resistance syndrome caused by two novel mutations in the NR3C1 gene. Endocrinol Nutr. 2016;63(7):369-371.
  93. Vitellius G, Fagart J, Delemer B, Amazit L, Ramos N, Bouligand J, et al. Three novel heterozygous point mutations of NR3C1 causing glucocorticoid resistance. Hum Mutat. 2016;37(8):794-803.
  94. Zhu HJ, Dai YF, Wang O, Li M, Lu L, Zhao WG, et al. Generalized glucocorticoid resistance accompanied with an adrenocortical adenoma and caused by a novel point mutation of human glucocorticoid receptor gene. Chin Med J (Engl). 2011;124(4):551-555.
  95. Sherlock M, Scarsbrook A, Abbas A, Fraser S, Limumpornpetch P, Dineen R, et al. Adrenal Incidentaloma. Endocr Rev. 2020;41(6):775-820.
  96. Karl M, Lamberts SW, Koper JW, Katz DA, Huizenga NE, Kino T, et al. Cushing's disease preceded by generalized glucocorticoid resistance: clinical consequences of a novel, dominant-negative glucocorticoid receptor mutation. Proc Assoc Am Physicians. 1996;108(4):296-307.
  97. Vitellius G, Trabado S, Hoeffel C, Bouligand J, Bennet A, Castinetti F, et al. Significant prevalence of NR3C1mutations in incidentally discovered bilateral adrenal hyperplasia: Results of the French MUTA-GR Study. Eur J Endocrinol. 2018;178(4):411-423.
  98. Nicolaides NC, Roberts ML, Kino T, Braatvedt G, Hurt DE, Katsantoni E, et al. A novel point mutation of the human glucocorticoid receptor gene causes primary generalized glucocorticoid resistance through impaired interaction with the LXXLL motif of the p160 coactivators: dissociation of the transactivating and transreppressive activities. J Clin Endocrinol Metab. 2014;99(5):E902-907.
  99. Kino T, Stauber RH, Resau JH, Pavlakis GN, and Chrousos GP. Pathologic human GR mutant has a transdominant negative effect on the wild-type GR by inhibiting its translocation into the nucleus: Importance of the ligand-binding domain for intracellular GR trafficking. J Clin Endocrinol Metab. 2001;86(11):5600-5608.
  100. Charmandari E, Kino T, Souvatzoglou E, Vottero A, Bhattacharyya N, and Chrousos GP. Natural glucocorticoid receptor mutants causing generalized glucocorticoid resistance: molecular genotype, genetic transmission, and clinical phenotype. J Clin Endocrinol Metab. 2004;89(4):1939-1949.
  101. Malchoff CD, Javier EC, Malchoff DM, Martin T, Rogol A, Brandon D, et al. Primary cortisol resistance presenting as isosexual precocity. J Clin Endocrinol Metab. 1990;70(2):503-507.
  102. Vottero A, Kino T, Combe H, Lecomte P, and Chrousos GP. A novel, C-terminal dominant negative mutation of the GR causes familial glucocorticoid resistance through abnormal interactions with p160 steroid receptor coactivators. J Clin Endocrinol Metab. 2002;87(6):2658-2667.
  103. Karl M, Lamberts SW, Detera-Wadleigh SD, Encio IJ, Stratakis CA, Hurley DM, et al. Familial glucocorticoid resistance caused by a splice site deletion in the human glucocorticoid receptor gene. J Clin Endocrinol Metab. 1993;76(3):683-689.
  104. Lin L, Wu X, Hou Y, Zheng F, and Xu R. A novel mutation in the glucocorticoid receptor gene causing resistant hypertension: A case report. Am J Hypertens. 2019;32(11):1126-1128.
  105. Charmandari E, Ichijo T, Jubiz W, Baid S, Zachman K, Chrousos GP, et al. A novel point mutation in the amino terminal domain of the human glucocorticoid receptor (hGR) gene enhancing hGR-mediated gene expression. J Clin Endocrinol Metab. 2008;93(12):4963-4968.
  106. Bouligand J, Delemer B, Hecart AC, Meduri G, Viengchareun S, Amazit L, et al. Familial glucocorticoid receptor haploinsufficiency by non-sense mediated mRNA decay, adrenal hyperplasia and apparent mineralocorticoid excess. PLoS One. 2010;5(10):e13563.
  107. Ruiz M, Lind U, Gåfvels M, Eggertsen G, Carlstedt-Duke J, Nilsson L, et al. Characterization of two novel mutations in the glucocorticoid receptor gene in patients with primary cortisol resistance. Clin Endocrinol (Oxf). 2001;55(3):363-371.
  108. Ma L, Tan X, Li J, Long Y, Xiao Z, De J, et al. A novel glucocorticoid receptor mutation in primary generalized glucocorticoid resistance disease. Endocr Pract. 2020;26(6):651-659.
  109. Trebble P, Matthews L, Blaikley J, Wayte AW, Black GC, Wilton A, et al. Familial glucocorticoid resistance caused by a novel frameshift glucocorticoid receptor mutation. J Clin Endocrinol Metab. 2010;95(12):E490-499.
  110. Vitellius G, Delemer B, Caron P, Chabre O, Bouligand J, Pussard E, et al. Impaired 11β-hydroxysteroid dehydrogenase type 2 in glucocorticoid-resistant patients. J Clin Endocrinol Metab. 2019;104(11):5205-5216.
  111. Raef H, Baitei EY, Zou M, and Shi Y. Genotype-phenotype correlation in a family with primary cortisol resistance: possible modulating effect of the ER22/23EK polymorphism. Eur J Endocrinol. 2008;158(4):577-582.
  112. Molnár Á, Patócs A, Likó I, Nyírő G, Rácz K, Tóth M, et al. An unexpected, mild phenotype of glucocorticoid resistance associated with glucocorticoid receptor gene mutation case report and review of the literature. BMC Med Genet. 2018;19(1):37.
  113. Donner KM, Hiltunen TP, Jänne OA, Sane T, and Kontula K. Generalized glucocorticoid resistance caused by a novel two-nucleotide deletion in the hormone-binding domain of the glucocorticoid receptor gene NR3C1. Eur J Endocrinol. 2013;168(1):K9-k18.
  114. Hurt DE, Suzuki S, Mayama T, Charmandari E, and Kino T. Structural analysis on the pathologic mutant glucocorticoid receptor ligand-binding domains. Mol Endocrinol. 2016;30(2):173-188.
  115. Cole TJ, Blendy JA, Monaghan AP, Krieglstein K, Schmid W, Aguzzi A, et al. Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation. Genes Dev. 1995;9(13):1608-1621.
  116. Nicolaides NC, and Charmandari E. Novel insights into the molecular mechanisms underlying generalized glucocorticoid resistance and hypersensitivity syndromes. Hormones (Athens). 2017;16(2):124-138.
  117. Kino T, Liou SH, Charmandari E, and Chrousos GP. Glucocorticoid receptor mutants demonstrate increased motility inside the nucleus of living cells: time of fluorescence recovery after photobleaching (FRAP) is an integrated measure of receptor function. Mol Med. 2004;10(7-12):80-88.
  118. Kaziales A, Rührnößl F, and Richter K. Glucocorticoid resistance conferring mutation in the C-terminus of GR alters the receptor conformational dynamics. Sci Rep. 2021;11(1):12515.
  119. Dahlman-Wright K, Wright A, Gustafsson JA, and Carlstedt-Duke J. Interaction of the glucocorticoid receptor DNA-binding domain with DNA as a dimer is mediated by a short segment of five amino acids. J Biol Chem. 1991;266(5):3107-3112.
  120. van Rossum EF, and Lamberts SW. Polymorphisms in the glucocorticoid receptor gene and their associations with metabolic parameters and body composition. Recent Prog Horm Res. 2004;59:333-357.
  121. Vitellius G, Trabado S, Bouligand J, Delemer B, and Lombès M. Pathophysiology of glucocorticoid signaling. Ann Endocrinol (Paris). 2018;79(3):98-106.
  122. Wester VL, Koper JW, van den Akker EL, Franco OH, Stolk RP, and van Rossum EF. Glucocorticoid receptor haplotype and metabolic syndrome: the Lifelines cohort study. Eur J Endocrinol. 2016;175(6):645-651.
  123. Ferriman D, and Gallwey JD. Clinical assessment of body hair growth in women. J Clin Endocrinol Metab. 1961;21:1440-1447.
  124. Marshall WA, and Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child. 1969;44(235):291-303.
  125. Marshall WA, and Tanner JM. Variations in the pattern of pubertal changes in boys. Arch Dis Child. 1970;45(239):13-23.
  126. Chrousos GP, Kino T, and Charmandari E. Evaluation of the hypothalamic-pituitary-adrenal axis function in childhood and adolescence. Neuroimmunomodulation. 2009;16(5):272-283.
  127. Kino T, Pavlatou MG, Moraitis AG, Nemery RL, Raygada M, and Stratakis CA. ZNF764 haploinsufficiency may explain partial glucocorticoid, androgen, and thyroid hormone resistance associated with 16p11.2 microdeletion. J Clin Endocrinol Metab. 2012;97(8):E1557-1566.
  128. Kadmiel M, and Cidlowski JA. Glucocorticoid receptor signaling in health and disease. Trends Pharmacol Sci. 2013;34(9):518-530.
  129. Chrousos GP, and Kino T. Glucocorticoid action networks and complex psychiatric and/or somatic disorders. Stress (Amsterdam, Netherlands). 2007;10(2):213-219.

 

Hypoparathyroidism and Pseudohypoparathyroidism

ABSTRACT

 

In primary hypoparathyroidism with hypocalcemia and hyperphosphatemia, deficient parathyroid hormone (PTH) secretion most commonly occurs from surgical excision of, or damage to, the parathyroid glands. The term idiopathic hypoparathyroidism describes isolated cases when a cause is not obvious, and there is no family history. However, hypoparathyroidism is also a feature common to a variety of hereditable syndromes that may present de novo. Familial isolated hypoparathyroidism may show autosomal dominant, autosomal recessive, or X-linked inheritance. Genes involved include PTH, SOX3, CASR, GNA11 and GCM2. Parathyroid hypoplasia is a frequent feature of 22q11.2 deletion syndrome with involvement of the TBX1 gene. The Hypoparathyroidism, Nerve Deafness, and Renal Dysplasia syndrome is due to haploinsufficiency of the GATA3 gene. Antibodies against parathyroid tissue are found in isolated hypoparathyroidism or combined with other endocrine deficiencies. Antibodies against the CASR occur in type 1 autoimmune polyglandular syndrome, due to mutations of the AIRE gene, or in acquired hypoparathyroidism. Disorders characterized by end-organ resistance to PTH are described collectively by the term pseudohypoparathyroidism (PHP), and PHP1A and PHP1B are caused by maternally-inherited changes at the imprinted GNAS complex gene that encodes the Gsα protein. Deleterious mutations of the PTH1R gene show resistance to PTH and PTHrP and present as Blomstrand lethal chondrodysplasia, Eiken syndrome, endochondromatosis, and primary failure of tooth eruption. Calcium and vitamin D are the standard therapy for the management of hypoparathyroidism, with hormone replacement [recombinant human PTH(1-84)] therapy recently becoming an option. Calcilytics, PTH analogs, and orally active small molecule PTH1R agonists may, in the future, join the treatment armamentarium.

 

PRIMARY HYPOPARATHYROIDISM

 

Primary hypoparathyroidism is caused by a group of heterogeneous conditions in which hypocalcemia and hyperphosphatemia occur as a result of deficient parathyroid hormone (PTH) secretion (1). This most commonly results from surgical excision of, or damage to, the parathyroid glands. However, autoimmune disease is also a significant factor in acquired cases, and genetic forms of hypoparathyroidism due to decreased PTH secretion are not rare (Table 1).

 

Table 1. Forms of Hypoparathyroidism having a Genetic Basis

   Isolated

         1) Autosomal dominant

                   A) PTH mutation

                   B) CASR activating mutation (ADH1)

                         a)  Bartter Syndrome Type V

                   C) GCM2 mutation (dominant negative)

                   D) GNA11 activating mutation (ADH2)

         2) Autosomal recessive

                   A) PTH mutation

                   B) GCM2 mutation

         3) X-linked

   Congenital multi-system syndromes*

         1) DiGeorge 1 (22q11) & 2 (10p)

         2) Barakat/HDR

         3) Kenny-Caffey 1 & 2 and Sanjad-Sakati

   Metabolic disease

         1) Mitochondrial neuromyopathies

         2) Long-chain hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency

         3) Heavy-metal storage disorders

   Autoimmune disease

         1) Autoimmune polyendocrine syndrome type I (APS-1 / APECED)

   Parathyroid resistance syndromes

         1) Pseudohypoparathyroidism

         2) Blomstrand chondrodysplasia and related PTH receptor defects

         3) Hypomagnesemia

* Clarke et al. (2) list other potential syndromic associations with hypoparathyroidism, including: CHARGE (Coloboma, Heart defect, Atresia choanae, Retarded growth and development, Genital hypoplasia, Ear anomalies/deafness), Dubowitz, lymphedema, nephropathy & nerve deafness

 

The signs and symptoms of hypoparathyroidism include evidence of latent or overt neuromuscular hyperexcitability due to hypocalcemia (Table 2). The effect may be aggravated by hyperkalemia or hypomagnesemia, but there is wide variation in the severity of symptoms. Patients may complain of circumoral numbness, paresthesias of the distal extremities, or muscle cramping, which can progress to carpopedal spasm or tetany. Laryngospasm or bronchospasm and seizures may also occur. Other less specific manifestations include fatigue, irritability, and personality disturbance. A comprehensive list of features associated with hypocalcemia can be found in the Endotext chapter, “Hypocalcemia: diagnosis and treatment” by Schafer & Shoback (3).

 

Severe hypocalcemia may be associated with a prolonged QTc interval on electrocardiography, which reverses with treatment. More extensive cardiomyopathic changes may be seen. These include chest pain, elevated enzymes (CPK), left ventricular impairment, and T-wave inversion, suggestive of a myocardial infarction (4, 5) . Patients with chronic hypocalcemia may have calcification of the basal ganglia or more widespread intracranial calcification, detected by skull X-ray or CT scan. Also seen are extrapyramidal neurological symptoms (more often with intracranial calcification), subcapsular cataracts, band keratopathy, and abnormal dentition.

 

Table 2. Some Clinical Features of Hypocalcemia

·       Neuromuscular irritability

·       Paresthesias

·       Laryngospasm

·       Bronchospasm

·       Tetany

·       Seizures

·       Chvostek sign

·       Trousseau sign

·       Prolonged QT interval on ECG

 

Increased neuromuscular irritability may be demonstrated by eliciting a Chvostek or Trousseau sign. A positive Chvostek sign is a prolonged reflex contraction of the facial muscle in response to a digital tap on the cheek just anterior to the ear. As with other hyperreflexias, up to 20% of normal individuals may demonstrate a slight positive reaction. A positive Trousseau sign is carpopedal spasm induced by inflation of a blood pressure cuff covering the upper arm to 20 mm Hg above systolic blood pressure for three minutes. This response reflects the heightened irritability of nerves undergoing pressure ischemia.

 

In hypoparathyroidism, serum calcium concentrations are decreased and serum phosphate levels are increased. Serum PTH is low or undetectable. (The important exception is PTH resistance, discussed further below.) Usually, serum 1,25-dihydroxyvitamin D (1,25(OH)2D) is low, but alkaline phosphatase activity is normal. Despite an increase in fractional excretion of calcium, intestinal calcium absorption and bone resorption are both suppressed. The renal filtered load of calcium is decreased, and the 24-h urinary calcium excretion is reduced; nephrogenous cyclic AMP excretion is low and renal tubular reabsorption of phosphate is elevated.

 

The terms idiopathic or isolated hypoparathyroidism have been traditionally used to describe isolated cases of glandular hypofunction when a cause is not obvious and there is no family history. However, hypoparathyroidism is a feature common to a variety of heritable syndromes that may present de novo. Hypoparathyroidism can occur because of a congenital hypoplasia/aplasia with or without other congenital anomalies such as dysmorphic facies, immunodeficiency, lymphedema, nephropathy, nerve deafness or cardiac malformation. Thus, in patients with hypoparathyroidism of uncertain onset, a careful examination of craniofacial features and assessment of endocrine, cardiac and renal systems should be performed to exclude a syndromic cause. Similarly, autoimmune hypoparathyroidism can occur as an isolated endocrine condition or with other glandular deficiencies in a pluriglandular autoimmune syndrome, requiring attention to multi-organ endocrine dysfunction.

 

A significant number of patients with idiopathic hypoparathyroidism and hypercalciuria, but no other anomalies may be found to have de novo activating mutations of the CASR gene.

 

Because of the implications for treatment, CASR molecular screening of patients with this presentation is recommended (6, 7).

 

Familial Isolated Hypoparathyroidism

 

Familial isolated hypoparathyroidism (FIH) may show autosomal dominant, autosomal recessive, or X-linked inheritance.

 

In a few instances of autosomal dominant disease, a mutation in the PTH gene (MIM# 168450 (8) - http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM) has been found. In one family, a missense mutation (C18R) in the signal sequence of the preproPTH precursor has been identified (9) and the mutant shown to be defective in vitro in processing preproPTH to proPTH, although, as patients had one normal gene copy, the autosomal dominant mode of inheritance remained unexplained. Then, further studies in transfected cells showed that the mutant was trapped in the endoplasmic reticulum (ER) promoting ER stress and apoptosis (10). In a family with autosomal recessive hypoparathyroidism, a different, homozygous, signal sequence mutation (S23P) segregates with affected status (11). This mutation may prevent proper cleavage of the signal peptide during processing of the nascent protein. In a girl with isolated hypoparathyroidism, a homozygous S23X signal sequence mutation was found predicting a truncated inactive PTH peptide (12). However, the circulating PTH level was not undetectable, suggesting some translational readthrough of the mutant preproPTH mRNA. A homozygous [Cys25]PTH(1-84) mutation that impairs PTHR1 activation was identified in an idiopathic hypoparathyroid family (13). Elevated circulating PTH levels were found in some (but not all) assays thus defining a novel form of hypoparathyroidism. In another family with autosomal recessive hypoparathyroidism, a donor splice site mutation at the exon 2/intron 2 junction of the PTH gene was identified (14). The mutation leads to exon skipping and loss of exon 2 containing the initiation codon and signal sequence of preproPTH mRNA. The SOX3 gene encodes a transcriptional factor likely involved in the embryonic development of the parathyroid gland (15). In two multigeneration families with X-linked recessive hypoparathyroidism exhibiting neonatal onset of hypocalcemia and parathyroid agenesis, the trait was mapped to a 906-kb region on distal Xq27 that contains three genes including SOX3 but no intragenic mutations were found (MIM# 307700). An interstitial deletion-insertion involving chromosomes 2p25.3 and Xq27.1 was found downstream of SOX3 and was speculated to exert a positional effect on SOX3 expression (16).

 

Gain-of-function mutations in the calcium-sensing receptor (CASR) gene (MIM#601199) have been identified in a number of families clinically diagnosed with autosomal dominant hypocalcemia type 1 (ADH1 – MIM#515361) (17, 18). In the parathyroid gland, the activated CASR suppresses PTH secretion, and in the kidney, it induces hypercalciuria that may contribute to the hypocalcemia. In many cases of ADH1, the family history is positive, but de novo mutations are quite common (19, 20). Mosaicism for de novo mutation in an otherwise healthy parent has been described (21), and may explain some cases of apparently recessive disease. Most importantly, there are implications for counseling parents about the risks of recurrence.

 

Almost all of the activating mutations are missense and appear almost equally divided between the amino-terminal third of the extracellular domain (ECD) and the transmembrane domain (TMD). Of special interest is the cluster of ECD mutations (A116T to C131W) that cause an increase in receptor sensitivity to extracellular calcium, suggesting that this region is critical for receptor activation. This cluster overlaps the two cysteine residues –cys-129 and cys-131– involved in the interface of the mature protein dimer (22). Further details can be found in the locus-specific database –http://data.mch.mcgill.ca/casrdb/ (23) and (24).

 

Although Bartter syndrome subtype V is represented by only a handful of cases with heterozygous severe activating mutations in the CASR (MIM#601199), it provides additional insight into the functioning of the CaSR in the thick ascending limb (TAL) of the nephron (25-27). Bartter syndrome encompasses a heterogeneous group of electrolyte homeostasis disorders, the common features of which are hypokalemic alkalosis, hyperreninemia, and hyperaldosteronism. Bartter syndrome subtypes I–IV are autosomal recessive disorders due to inactivating mutations in the following ion transporters or channels active in the TAL: type I, the sodium potassium-chloride cotransporter (NKCC2); type II, the outwardly rectifying potassium channel (ROMK); type III, the voltage-gated chloride channel (CLC-Kb); type IV, Barttin, a chloride channel beta-subunit that is required for trafficking of CLC-Ka and CLC-Kb. Patients with the autosomal dominant Bartter syndrome subtype V have, in addition to the classic features of the syndrome, hypocalcemia, and may exhibit neuromuscular manifestations, seizures, and basal ganglia calcifications. NKCC2 and ROMK in the apical membrane (luminal side) of the TAL have been proposed to generate a transepithelial electrochemical gradient that drives passive paracellular transport of Na+, Mg2+, and Ca2+ from the lumen to blood (28). The CASR is situated in the basolateral membrane (antiluminal side) and, when activated, increases 20-hydroxyeicosatetraenoic acid and decreases cAMP concentrations, both of which would inhibit ROMK and NKCC2 activities (28, 29). Thus, severe activating mutations of the CASR lead to the salt wasting of Bartter syndrome in addition to the hypercalciuric hypocalcemia of ADH1.

 

Heterozygous gain-of-function missense mutations of GNA11 have been identified in ADH patients without detectable CASR activating mutations (30-33). The GNA11 activating mutations increase the sensitivity of the parathyroid gland and renal tubule to extracellular calcium concentrations. Autosomal dominant hypocalcemia and hypoparathyroidism due to CASR and GNA11 mutations are now designated as ADH type 1 (MIM#601198) and type 2 (MIM#615361) respectively. The human Gα11 protein (a Gq family member – MIM#139313) has 359 amino acids with an α-helical domain in the NH2-terminal region, a GTPase domain in the COOH-terminal region, and three switch regions (SR1-3) in the middle portion that change conformation based on whether GTP or GDP is bound (34). The R80C, R181Q, S211W, F341L, and V304M mutations found in hypocalcemic individuals are predicted by 3D modeling to alter the normal Gα11 protein structure. Moreover, cells stably expressing the CASR and transfected with the mutants exhibit increased sensitivity to changes in extracellular calcium (30-33).

 

Inactivating mutations in the CASR regulator, the adaptor protein 2 sigma subunit encoded by the AP2S1 gene, cause familial hypocalciuric hypercalcemia type 3 (35). The search for activating mutations in AP2S1 in familial and sporadic isolated hypoparathyroid patients negative for CASR or GNA11 mutations that would represent an additional genetic cause of ADH has thus far been negative (36, 37).

 

Recessively inherited FIH may occur with mutations of the glial cells missing-2 gene (GCM2; MIM#603716). The GCM2 gene localizes to chromosome 6p24.2 and encodes a transcription factor. It is expressed in the PTH-secreting cells of the developing parathyroid glands and is critical for their development in terrestrial vertebrates (38-40). A patient with neonatal hypoparathyroidism was found to be homozygous for a partial deletion acquired from both parents (41), and a pair of siblings with homozygous mutations has been reported (42). Additional studies have identified inactivating GCM2 mutations in cases with autosomal recessive FIH (43, 44). On the other hand, heterozygous mutations that cause dominant-negative GCM2 mutants have also been identified in patients with autosomal dominant hypoparathyroidism (43, 45, 46).  Additional recessive and dominant GCM2 mutations have been noted in this gene that continues to be expressed in the adult parathyroid [see (47)]. Nevertheless, it appears that the prevalence of genetic defects affecting GCM2 function is not high in isolated hypoparathyroidism, as a recent study investigating 20 unrelated cases with this disorder (10 familial and 10 sporadic) failed to identify any GCM2 mutations segregating with the disease and/or leading to loss of function (48). Of further interest is that a genetic variant, Y282D that demonstrates significantly enhanced transcriptional activity relative to wild-type GCM2 associates with hyperparathyroidism in some cohorts of the sporadic primary disorder (49). Most recently, novel heterozygous active GCM2 variants that segregate with affected status in some kindreds with familial isolated hyperparathyroidism have been described (50). Thus, like CASR and GNA11, both gain-of-function and loss-of-function variants of GCM2 may contribute to calcemic disorders.

 

Hypoparathyroidism with Syndromic Features

 

Hypoparathyroidism due to parathyroid hypoplasia is a frequent feature of 22q11.2 microdeletions, the most common cause of DiGeorge syndrome 1 (DS1; MIM#188400) (51, 52) . This syndrome complex arises from a failure of the third and fourth pharyngeal pouches to develop, leading to agenesis or congenital hypoplasia of the parathyroid glands, thymus, and the anterior heart field. Patients with DS1 may typically present with neonatal hypocalcemic seizures due to hypoparathyroidism, severe infections due to thymic hypoplasia, and conotruncal heart defects (53). Because a microdeletion is involved, the identification of novel developmental genes in the 22q11 region has been keenly pursued. One of the genes is TBX1, encoding a DNA-binding transcription factor of the T-box family known to have important roles in vertebrate and invertebrate organogenesis and pattern formation (54, 55). Mouse models with Tbx1 haploinsufficiency established the essential contribution of this factor to conotruncal development (56), and placed it in developmental context during organogenesis (57, 58). However, while the Tbx1 null mutant mice had all the developmental anomalies of DS1 – thymic and parathyroid hypoplasia, abnormal facial structures and cleft palate, skeletal defects and cardiac outflow abnormalities – Tbx1 haploinsufficiency in mice was associated with only defects of the fourth pharyngeal pouch responsible for the cardiac outflow abnormalities (59). cDNA microarray analyses of mice lacking Tbx1 have identified Gcm2 as one of the downregulated genes in the pharyngeal region, indicating that Tbx1 is upstream of Gcm2 (60). Furthermore, as Tbx1 is regulated by sonic hedgehog (Shh) (61), a Shh-Tbx1-Gcm2 parathyroid developmental pathway is indicated.

 

The basis for the phenotypic differences between DGS1 patients who are heterogeneous for TBX1 loss and the Tbx1+/- mice is unclear but could reflect a species-specific gene dosage requirement together with roles of downstream genes regulated by Tbx1. Some patients may have late-onset DGS1 and develop symptomatic hypocalcemia in childhood or later with only subtle phenotypic abnormalities (62, 63). Of note is that the age of diagnosis in rare families with DGS1 patients having TBX1 inactivating (missense or frameshift) mutations ranged from 7 to 46 years in keeping with late-onset DGS1 (54).

 

The 22q11.2 deletion syndrome (22q11.2DS) encompasses a wider spectrum of clinical conditions that includes isolated congenital heart disease and velocardiofacial (VCF) syndrome (52). Associated craniofacial abnormalities include cleft palate, pharyngeal insufficiency and mildly dysmorphic facies. In the VCF syndrome, anatomical anomalies of the pharynx are prominent and hypernasal speech due to abnormal pharyngeal musculature with or without cleft palate is typical. In most patients, some degree of intellectual deficit is present and there is strong predisposition to psychiatric illness (schizophrenia or bipolar disorder) in adolescents and adults (64, 65). Further information, both clinical and educational, can be found at web sites specifically devoted to this condition [see (66)].

 

The 22q11.2DS is due to one of the most common microdeletions (1 in 4000 live births), and it may go clinically unrecognized in its milder or variant forms. Most cases with hypoparathyroidism (~50% of cases) are the result of de novo deletion through meiotic non-allelic homologous recombination, and driven by a unique cluster of low copy repeats designated LCR22 A-H [see (66, 67) ]. Most commonly (~85% of cases), a deletion of ~3 Mb is found, encompassing proximal repeats A to D. Many of the others (~10% of cases) involve atypical nested deletions including those spanning LCR22 A to B. Thus, LSR22 A to B, which includes the TBX1 gene, is the primary site contributing to parathyroid dysgenesis. Detailed characterization and long-term follow-up for the hypoparathyroid component of this disorder is ongoing.

 

Although most cases of DiGeorge syndrome are sporadic, as mentioned above autosomal dominant inheritance is not unknown. In utero influences may be important determinants of the clinical picture, since there are instances of monozygotic twins with discordant phenotypes (68-70). Phenocopies occur with diabetic embryopathy, fetal alcohol syndrome, and retinoid embryopathy. In rare instances, it has been shown that a phenotypically normal parent can transmit a microdeletion to an offspring. Such parents have been found to carry a duplication of the 22q11 on the second chromosome, and the combination of duplication and deletion alleles in a parent generates a balanced state, termed “gene dosage compensation” (71, 72).

 

Although the hypoparathyroidism affects about half of all carriers, it is usually not severe, and frequently treatment following neonatal hypocalcemia can be tapered or stopped in older children. However, the hypoparathyroidism may also remain asymptomatic until adolescence or emerge at times of stress, such as corrective cardiac surgery or severe infection, suggesting that continued surveillance of parathyroid gland reserve is important (73-75). 

 

Traditionally, diagnosis of 22q11.2DS is established with specific cytogenetic studies -- usually with locus-specific fluorescence in-situ hybridization (FISH) testing. These tests will pick up many of the larger common deletions that involve regions of low-copy number repeats (LCRs). However, specific chromosomal array-based and MLPA analyses are now preferred, as they have been shown to have increased sensitivity for smaller deletions (66). Recently, the diagnostic power of next-generation sequencing has been harnessed to identify almost all of the microdeletions underlying sporadic and inherited forms of the disorder (52). Non-invasive prenatal screening and pre-implantation genetic diagnosis) are also clinically available (76). Because the clinical picture is so variable and the prevalence so high, testing for 22q11.2 microdeletion should be considered in the workup for any new hypoparathyroid case for which another cause is not found. Finally, distinct genetic defects can coexist with 22q11.2DS, as exemplified by the finding of concurrence of this syndrome in an adolescent with longstanding hypercalcemia who had familial hypocalciuric hypercalcemia type 3 due to an AP2S1 mutation (77).

 

Clinicians will also want to be aware that a small but significant minority (~10%) of patients will have associated autoimmune disease, driven in part, perhaps, by the thymus-based defect in T cell function (64,79). Among the more common (non-endocrine) conditions are arthritis, celiac disease, and autoimmune hematologic disease, particularly idiopathic thrombocytopenic purpura. Autoimmune thyroid disease, with either hypo- or hyperparathyroid states, has been reported (78, 79), and serum TSH assay should be measured regularly. It has been suggested that the later-onset hypoparathyroid disease may be partly autoimmune in origin, not developmental. A survey of 59 Norwegian patients showed discordance of adult onset disease with neonatal hypoparathyroidism, but a significant correlation with parathyroid autoantibodies and the presence of autoimmune disease (78).

 

The clinical features of DiGeorge syndrome, including hypoparathyroidism, also occur with other cytogenetic abnormalities, notably chromosome 10p haploinsufficiency (80, 81). Deletions of two non-overlapping regions of chromosome 10p contribute to DiGeorge syndrome 2; DS2 at 10p13-14 (82), and the Barakat or HDR (Hypoparathyroidism, Nerve Deafness, and Renal Dysplasia) syndrome (MIM#146255) (83, 84) at 10p14-10pter (85, 86). The latter is due to haploinsufficiency of GATA3 (MIM#131320), which encodes a dual zinc finger transcription factor (87) that is essential for normal embryonic development of the parathyroids, auditory system, and kidney. Since the original description, several additional GATA3 loss-of-function mutations have been described in HDR patients [e.g., (88-91)]. Heterozygous Gata3-deficient mice develop parathyroid abnormalities as revealed by challenge with a diet low in calcium and vitamin D that are due to dysregulation of the parathyroid-specific transcription factor, Gcm2. Gata3-/- embryos at E12.5 lack Gcm2 expression and have gross defects in the fourth pharyngeal pouches, including absent parathyroid/thymus primordia (92). GATA3 transactivates the GCM2 promoter and, with GCM2, forms part of a transcriptional cascade essential for the differentiation and survival of parathyroid progenitor cells. 

 

In another congenital disorder, Kenny-Caffey syndrome, hypoparathyroidism is found variably associated with the typical picture of growth retardation, osteosclerosis, cortical thickening of the long bones, and delayed closure of the anterior fontanel (93-96). The original description of the syndrome was of the autosomal dominant form now identified as KCS-2 (MIM#127000) that is caused by heterozygous mutations in the FAM111A gene (97-99). The full functions of FAM111A and how mutations in it cause the disorder are unclear. FAM111A has some homology to peptidases, and is involved with chromatin structure during DNA replication (100). KS-2 is allelic to the lethal disorder, osteocraniostenosis (OCS, MIM#6023611). Hypocalcemia due to hypoparathyroidism was found in some OCS patients who survived the perinatal period (96).

 

A recessively inherited form of Kenny-Caffey syndrome (KCS-1, MIM#244460) was noted to be similar to the recessive Sanjad-Sakati syndrome (MIM#241410) characterized by congenital hypoparathyroidism, seizures, growth and developmental retardation and characteristic dysmorphic features, including deep set eyes, depressed nasal bridge with beaked nose, long philtrum, thin upper lip, micrognatia and large, floppy ear lobes. Radiographs showed medullary stenosis reminiscent of Kenny-Caffey syndrome (96, 101). Linkage studies localized the recessive KCS-1 and Sanjad-Sakati syndromes to 1q42-43, and causative mutations in the tubulin chaperone E, TBCE, gene were identified in what is now known as Hypoparathyroidism, Retardation and Dysmorphism (HRD) syndrome (96, 102, 103) . This highlighted the role of TBCE that binds microtubules and proteasomes and protects against misfolded stress (104) in parathyroid development (105).

 

Hypoparathyroidism due to Metabolic Disease

 

Hypoparathyroidism is also a variable component of the neuromyopathies caused by mitochondrial gene defects (106). Among these are the Kearns-Sayre syndrome (ophthalmoplegia, retinal degeneration, and cardiac-conduction defects) (MIM#530000), the Pearson marrow pancreas syndrome (lactic acidosis, neutropenia, sideroblastic anemia, and pancreatic exocrine dysfunction) (107) (MIM#557000) and mitochondrial encephalomyopathy (MIM#540000). The molecular defects range from large deletions and duplications of the mitochondrial genomes in a large number of tissues (108, 109) to single base-pair mutations in one of the transfer RNA genes found only in a restricted range of cell types (MIM#590050). The role of these mitochondrial mutations in the pathogenesis of hypoparathyroidism remains to be clarified. However, mutations in HADHB, that encodes the β-subunit of mitochondrial trifunctional protein, cause infantile onset hypoparathyroidism and peripheral polyneuropathy (110).

 

Long-chain hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency (MIM#600890)
is an inborn error of oxidative fatty acid metabolism that may be accompanied by hypoparathyroidism (111). Whether the parathyroid disease is directly related to the enzyme deficiency or secondary to the accompanying mitochondrial disease needs further study.

 

Parathyroid insufficiency and symptoms of hypocalcemia are occasionally seen in inherited metabolic disorders leading to excess storage of iron (thalassemia, Diamond-Blackfan anemia, hemochromatosis) or copper (Wilson disease)(112). In most instances, there is similar dysfunction in other endocrine glands, and the parathyroid disease is usually mild. Nonetheless, recognition of the hypoparathyroid state may help explain otherwise non-specific symptoms and aid in overall management of these multisystem diseases.

 

Autoimmune Hypoparathyroidism:  Acquired and Inherited Disorders

 

Antibodies directed against parathyroid tissue have been detected in up to 38% of patients with isolated hypoparathyroid disease, and over 40% of patients having hypoparathyroidism combined with other endocrine deficiencies (113, 114).  Subsequently, a survey of a parathyroid expression library led to the identification of one protein selectively associated with the autoimmune process, the NACHT leucine-rich-repeat protein 5 (NALP5). Elevated antibody titers occur in half the patients with autoimmune hypoparathyroidism, with or without another autoimmune disease, but uncommonly in other conditions without hypoparathyroidism (114, 115).

 

Antibodies against the extracellular domain of the parathyroid CASR were originally reported in more than half of patients with either type 1 autoimmune polyglandular syndrome (APS-1, also known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy or APECED), MIM# 240300, (116) or acquired hypoparathyroidism associated with autoimmune hypothyroidism (117). This finding was confirmed in a subsequent study of 51 cases of idiopathic hypoparathyroidism, but there was a 13% positive rate in controls (118). Other studies of APS-1 patients have also identified elevated CASR antibodies in some cases but at a lower frequency (119-121). Although some have suggested that CASR antibody assays are clinically indicated in acquired hypoparathyroidism (122), it remains to be seen whether the autoantibodies are of primary or secondary importance (114, 123). There is now good evidence that autoantibodies can be functional activators of CASR and thereby could induce hypoparathyroidism. While presently there may not be a convenient clinical test for this, patient sera have been demonstrated to activate the CASR transfected into HEK cells in vitro (124). In some hypoparathyroid patients, both autoimmune parathyroid destruction and suppression by CASR activation may co-exist (125).

 

In APS-1, the most common associated manifestations are hypoparathyroidism with mucocutaneous candidiasis and Addison's disease. Additional features include pernicious anemia, chronic active hepatitis, alopecia, keratitis, gonadal failure, thyroid disease, pancreatic insufficiency, and diabetes mellitus (116). The phenotype is highly variable and patients may not express all elements of the basic triad, leading to the suggestion that the criteria used for molecular screening be relaxed (125, 126). The disease usually presents in infancy with chronic oral thrush, followed by hypoparathyroidism in the first decade, and then adrenocortical failure in the third. Interestingly, there is nearly 100% penetrance of hypoparathyroidism in females, but less than 60% in males, even though the adrenal hypofunction affects both sexes equally (119). Moreover, patients who develop the adrenal hypofunction first are less likely to be male and may never develop hypoparathyroidism. The responsible gene, called the autoimmune regulator (AIRE), maps to chromosome 21q22 and encodes a transcriptional regulator (127-129) . In the absence of AIRE protein, tissue-specific self-antigens are not expressed in the thymus and multiorgan autoimmunity develops, because negative selection of the T cells bearing the autoantigens is disrupted (130). Many patients with APS-1 can be shown to have autosomal recessive inheritance of the AIRE defect. In families with autosomal recessive mutations of AIRE, obligate heterozygotes may also have common autoimmune disorders but APECED is not seen (131). A phenocopy leading to acquired APS-1 may occur when the AIRE gene is silenced by thymic neoplasia (132). APS-1 has been associated with more than 300 mutations of the AIRE gene, and updates can be found in the online mutation database (https://grenada.lumc.nl/LOVD2/mendelian_genes/home.php?select_db=AIRE).

 

PARATHYROID RESISTANCE SYNDROMES

Pseudohypoparathyroidism

 

Several clinical disorders characterized by end-organ resistance to PTH have been described collectively by the term pseudohypoparathyroidism (PHP). They are associated with hypocalcemia, hyperphosphatemia, and increased circulating PTH. Target tissue unresponsiveness to the hormone manifests as a lack of increased phosphate excretion and, in some cases, cAMP excretion in response to PTH administration (133). The biochemical characteristics of the different forms of PHP are contrasted with those of hypoparathyroidism in Table 3.

 

Table 3. Biochemical Characteristics of Hypoparathyroidism and Pseudohypoparathyroidism

Defects

Serum PO4

PTH

25(OH)D

1,25(OH)2D

UcAMP*

UPO4*

Multiple Endocrine Defects

Hypoparathyroidism

-

-

-

Yes/No**

Pseudohypoparathyroidism

     Type 1a

-

Yes

     Type 1b

-

No/Yes#

     Type 1c

-

Yes

     Type 2

-

-

No

↑, increased; ↓, decreased; -, normal;

*Response to PTH infusion

**, depending upon the etiology.

#, variable, mild defects of the thyroid axis due to TSH resistance may be seen.

 

Albright Hereditary Osteodystrophy

 

Fuller Albright first recognized that the likely cause of the hypoparathyroid state in PHP is a constitutive absence of target tissue responsiveness (134). In many patients, the end-organ resistance is accompanied by a specific pattern of physical findings, called Albright hereditary osteodystrophy (AHO; MIM#300800). Typically, patients have short stature, round facies, brachydactyly, obesity, and ectopic soft tissue or dermal ossification(s) (osteoma cutis) (Figure 1). In the calvaria, this may manifest as hyperostosis frontalis interna (135). Intracranial calcification(s), cataracts and band keratopathy, subcutaneous calcifications, and dental hypoplasia are also common but are likely the consequences of longstanding hypoparathyroid hypocalcemia (Table 4, see below Figure 1). The brachydactyly may be asymmetric or not, and may involve one or both hands or feet, but the pattern is quite distinctive (136, 137). The shortening tends to involve the first distal phalanx, with a thumbnail (or first toenail) that is wider than it is long. The fourth and fifth metacarpals (or metatarsals) are frequently shortened out of proportion to the others and the second metacarpal is often spared. Radiographic analysis of the hands (pattern profiling) may be helpful in assessment of the brachydactyly (Figure 1)(138).

 

Figure 1. Albright’s hereditary osteodystrophy (AHO) and characteristic skeletal abnormalities. A) A child with AHO exhibiting short stature, obesity, and round facies. B) The hand X-ray of a patient with AHO, displaying brachydactyly of the fourth and fifth metacarpal bones. C) Dimpling over the knuckles of a clenched fist (also known as Archibald sign), indicating the short metacarpals. D) Evidence of brachydactyly in the hand, reflecting the shortened fourth and fifth metacarpals and the distal phalanx of the thumb. Images are from: Levine, MA (139).

 

Table 4. Incidence of signs and symptoms in PHP with AHOa

 

Percentage

Short stature

80

Obesity

50

Craniofacial

Round face

92

bLenticular opacities

44

Strabismus

10

bDental hypoplasia

51

bBasal ganglia calcification

50

Thickened calvaria

62

Mental deficit

75

Brachydactyly

Brachymetacarpia

68

Brachymetatarsia

43

Brachyphalangia

50

Other connective tissue features

Decreased bone density

15

Ectopic ossification

56

bSubcutaneous calcification

55

a Taken from Drezner and Neelon (1995).

b Features common to other forms of chronic hypoparathyroid hypocalcemia.

 

Although affected patients are generally short as adults, their bone age as children may be advanced and growth accelerated (138). Patients with AHO may be predisposed to hypertension (140), conductive and sensorineural hearing loss (135, 141), cord compression due to spinal anomalies (142), and movement disorders due to basal ganglia calcification (143). The features of AHO may be subtle in infancy or early childhood; in a few, there is little to see even in adulthood. The round facies, short neck, and low, flat nasal bridge are often accompanied by central obesity (144). A study showed that the obesity phenotype occurs primarily in those patients who also have multiple hormone resistance, i.e., PHP1A (see below), and according to data from mice, a hypothalamic mechanism, rather than hypothyroidism, is the primary underlying cause (145, 146). Interestingly, a study showed that GNAS mutations are not uncommon in severe childhood-onset obesity in the absence of other typical PHP findings (147).

 

Patients with brachydactyly, mental retardation, and other features closely resembling AHO have been found to carry microdeletions of chromosome 2q37; brachydactyly-mental retardation, BDMR; MIM#600430 (148). Genes important for skeletal and neurological development lie within this region. Haploinsufficiency of HDAC4 (MIM#605314), encoding a histone deacetylase that regulates gene expression during the development of many tissues including the bone, is responsible for the brachydactyly and the mental retardation in those patients (149). Isolated brachydactyly type E (BDE, MIM#113300) has been associated in sporadic cases with mutations in HOX13 (MIM#168470) (150) and mutations in the PTHLH gene (MIM#168470) on 12p11.2 that encodes PTHrP have been implicated. In one family with autosomal BDE a cis-regulatory site downregulates PTHLH in translocation t(8;12)(q13;p11.2) and downregulates its targets ADAMTS-7 and ADAMTS-12 leading to impaired chondrogenic differentiation (151). Affected individuals of one large family with BDE, short stature, and learning difficulties had an ~900 bp microdeletion encompassing PTHLH (152). Additional individuals with BDE and short stature from other different kindreds were found to have PTHLH missense, nonstop, and nonsense mutations (152). Different translocations affecting chromosome 12p have also been identified in two families with BDE, leading to increased abundance of a long noncoding RNA on chromosome 12q, which regulates the expression of PTHLH in cis and of the SOX9 gene located on chromosome 17q in trans (153). BDE is associated with hypertension in some cases, in which the disease is inherited in an autosomal dominant manner (termed HTNB). Missense mutations in PDE3A, a gene encoding a cAMP/cGMP phosphodiesterase, have been recently found in several unrelated families with HTNB. These mutations cause increased cAMP hydrolytic activity and thus lead to diminished cAMP signaling (154). Some patients with AHO-like features have been described, who also showed platelet Gs hypofunction. Those patients were found to have IGF2 hypermethylation and SNURF hypomethylation, as well as imprinting defects within GNAS, the gene encoding the stimulatory G protein alpha-subunit (Gsα; see below) (155).

 

PHP1A

 

PHP1A patients are characterized by AHO, PTH resistance, and evidence of target organ resistance to other hormones. Patient-derived cells are found to have a reduction in the activity of the Gsα subunit, which is part of the membrane-associated heterotrimeric stimulatory G-protein complex - transducing signals between G-protein coupled receptors and adenylate cyclase (156-158). Adenylyl cyclase catalyzes the synthesis of the second messenger cAMP, and therefore, PHP1A patients tend to have a deficiency in cAMP generation, particularly in certain tissues. As explained above, this deficiency is clear when measuring cAMP excretion in response to PTH administration.

 

The GNAS gene (MIM#168470) encoding the Gsα protein maps to 20q13.2-13.3 and has at least 4 alternative transcriptional start sites (Figure 2) and an antisense transcript, GNAS-AS1 (159). The three upstream exons and the preceding promoter regions are genetically imprinted, i.e., methylated in an allele specific manner. The promoter of the Gsα transcript, which uses exon 1, is unmethylated. Unlike the other alternative GNAS products, Gsα expression is biallelic except in a small set of tissues, where Gsα is derived predominantly from the maternal allele (160-164) . This tissue-specific monoallelic Gsα expression affects the penetrance of the PHP phenotype. The maternal transmission of the hormone resistance in PHP1A (165) can be explained by the silencing of the paternal Gsα allele, which would otherwise allow expression of 50% of Gsα protein (166). Thus, the full expression of a coding GNAS mutation, which occurs in maternally transmitted cases, leads to AHO plus hormone resistance (PHP1A). On the other hand, if the same mutation is inherited paternally, it causes AHO alone. The latter is termed pseudopseudohypoparathyroidism (PPHP). Thus, PHP1A and PPHP can be found in the same families. Note that a systematic nomenclature and classification, “inactivating PTH/PTHrP signaling disorder” (iPPSD), has been suggested for PHP1A, PPHP, and related disorders arising from abnormal PTH and/or cAMP signaling, accounting for the underlying genetic/epigenetic abnormalities and associated phenotypes (167).

 

Despite clinical evidence supporting imprinting in portions of the kidney tubule, it has been difficult to confirm this experimentally in humans (168). The imprinting of GNAS is complex and involves multiple differentially methylated regions (DMR) (159). Moreover, it is tissue-specific and may vary with developmental stage, although key imprinting of the A/B (also referred to as 1A) DMR is thought to be a primary event that occurs during gametogenesis and is maintained thereafter (169). Ablation of the Gsα ortholog in mice (Gnas) has confirmed that maternal, but not paternal, transmission of the deleted allele results in PTH resistance. The homozygous deletion of Gnas is embryonic lethal (160). Comparison of Gsα expression in mice with maternally vs paternally disrupted Gsα expression also demonstrated that Gsα expression is predominantly maternal in the renal cortex, but not in renal medulla (160, 170) . PTH resistance is delayed until after infancy in most PHP1A patients, and a study using mice demonstrated that the silencing of the paternal Gsα allele develops postnatally (171).

 

Figure 2. Simplified view of the GNAS region and its transcripts. The normal allele-specific methylation and expression patterns of the four alternate first exons of GNAS which splice onto exon 2 to produce transcripts encoding NESP55, XLαs, 1A (referred to as A/B in humans), and Gsα (which uses exon 1). NESP55 and XLαs promoters are oppositely imprinted: NESP55 is expressed from the maternal allele and its promoter region is methylated on the paternal allele, whereas XLαs is expressed from the paternal allele and its promoter is methylated on the maternal allele. Gsα is paternally silenced in some tissues e.g., renal proximal tubule cells, indicated by the dashed arrow. NESP55 protein is unrelated to Gsα, and its entire coding region is located within its first exon. In contrast, XLαs and Gsα proteins have identical COOH-terminal domains (encoded by exons 2-13), while their unique NH2-terminal domains are encoded within their respective first exons. Exon A/B (1A) does not have a translational start site but is transcriptionally active. Loss of exon A/B imprinting (methylation) is associated with decreased Gsα expression in renal proximal tubules and some other hormone-responsive tissues and is the typical cause of PHP1B. (figure from Liu et al., 2000, with permission).

 

A variety of inactivating mutations in the Gsα-coding portion of the GNAS gene have been identified in PHP1A patients (172, 173). The spectrum includes missense mutations, point mutations impairing efficient and accurate splicing, and small insertion/deletion mutations. The 4-bp deletion in exon 7 (DGACT 188/190) has been observed in multiple unrelated cases, suggesting that this may be a hot spot (174, 175). Several other mutations have also been observed in more than one kindred, indicating that additional susceptibility regions may exist. The identification of de novo germline mosaicism (176) is consistent with the view that most sporadic cases harbor new mutations, but the separation of such sporadic cases from familial ones, in which there is suppression of phenotype due to imprinting, may be difficult without detailed molecular studies.

 

PHP1A cases have been described in which no mutations of the GNAS gene have been found by nucleotide sequence analysis of exons encoding Gsα. This may be because the mutation is in a regulatory region of the gene not yet examined, or it may be that a large deletion prevents amplification of the mutant allele for subsequent analyses. In cases without identified GNAS coding mutations, an assessment of Gsα bioactivity in erythrocytes is helpful in ruling out regulatory region mutations or large deletions. A 35-kb deletion spanning exons 1 through 5 has been identified by using comparative genome hybridization in a patient with PHP1A in whom coding mutations had been ruled out, but a marked reduction of erythrocyte Gsα activity demonstrated (177).

 

Typically, PHP1A is associated with multiple hormone resistance, including thyroid stimulating hormone (TSH) and gonadotropins, causing hypothyroidism and gonadal failure, respectively. Because the hypothyroidism may express before hypocalcemia is observed (178), early surveillance of thyroid function is warranted. However, thyroid replacement from birth does not appear to prevent the mental deficit typical of PHP1A. In women, the hypogonadism is partial (179), and thus, oral contraceptives may help regulate the menstrual cycle. Estrogen can antagonize bone resorption, leading to an exacerbation of hypocalcemia (180), but placental 1,25-dihydroxyvitamin D synthesis likely obviates this effect altogether in pregnancy so women are frequently normocalcemic at that time (181). Abnormalities of the somatotropin axis have also been reported, with documentation of subnormal growth hormone release following stimulation by L-arginine or growth hormone-releasing hormone (182, 183).

 

The tissue-specific silencing of the paternal Gsα allele also plays a key role in the development of the additional hormone resistance phenotypes, as monoallelic Gsα expression has been demonstrated in the thyroid, the ovaries, and the pituitary (161-164). Studies have revealed that obesity also develops primarily in patients who inherit the inactivating Gsα mutations from their mothers (184). Gsα is not imprinted in the white adipose tissue (185), but the investigations of mice in which Gsα is ablated conditionally in the brain showed that Gsα is also monoallelic in certain parts of the hypothalamus (145, 146), thus explaining the imprinted mode of inheritance of the obesity phenotype. This likely reflects impaired signaling downstream of the melanocortin receptor type-4 (MC4R), given that it signals via G proteins including Gsα and that inactivating MC4R mutations are causal for dominantly inherited morbid obesity (186, 187). Indeed, almost all GNAS mutations identified in a large cohort of children with severe obesity impaired MCR4 signaling in cell-based assays (147). In mice, ablation of the maternal but not paternal Gnas allele in the dorsomedial nucleus of the hypothalamus leads to obesity (145), similar to the findings in mice with the conditional MC4R deficiency in this part of the brain (188). Like obesity, it has been noted that cognitive impairment, a typical AHO feature, also develops primarily after maternal inheritance of the inactivating Gsα mutation (189), although the underlying mechanisms behind the parental-specific inheritance of this phenotype have yet to be defined.

 

PHP1B

 

PHP1B is typically not associated with AHO or a generalized reduction in Gsα expression (190-192). PHP1B patients show a defect in renal PTH signaling, but an apparently normal response to PTH in bone (193, 194). Affected individuals are therefore functionally hypoparathyroid but show normal skeletal architecture and development. Due to unimpaired PTH responsiveness in bone, however, signs of hyperparathyroid bone disease (osteitis fibrosa cystica) are occasionally observed, complicating the picture (195). Biochemical abnormalities suggestive of thyroid stimulating hormone resistance are also seen in some patients (164). In fact, sometimes, PHP1B cases can present first with hypothyroidism (196, 197). A study also demonstrated short stature and growth hormone deficiency in monozygotic twins with PHP1B (198). Abnormalities of renal uric acid handling have been documented (199, 200). However, clinically significant hormone resistance is restricted to PTH in most cases. Because the hormone resistance is mostly limited to PTH, it was thought at one time that these findings could be explained by a defect in the type-1 parathyroid hormone receptor (PTH1R, MIM#168468). Sequence analyses, however, found no mutations in protein-coding exons or gene promoter regions of the gene (201-203), and studies of PHP1B families show no linkage to PTHR1 (204, 205).

 

Most cases of PHP1B are sporadic, but a familial form of PHP1B with an apparent autosomal dominant mode of inheritance also exists (AD-PHP1B). In four AD-PHP1B kindreds, linkage to chromosome 20q13.3 was established, the same region which includes the GNAS locus (204). In these families, the pattern of transmission suggested paternal imprinting, and inheritance is therefore the same as for PHP1A. A further 13 PHP1B subjects were studied, some of whom had bone responsiveness to PTH (166). All lacked methylation of the alternate exon A/B, an epigenetic defect that is postulated to inhibit expression of the functional exon 1-containing Gsα transcript in renal tissues only (Figure 2). Thus, the loss of methylation of the maternal exon A/B allele leads to the silencing of the maternal as well as paternal Gsα allele, causing PTH resistance specifically in renal proximal tubule cells. A genetic analysis indicated that mutations in a regulatory region some distance from the GNAS coding exons were likely to account for the unique imprinting defect(s) associated with PHP1B (206). A search for the mutation revealed the presence of a 3 kb microdeletion that segregated with the disease in 12 kindreds with AD-PHP1B and also occurred in 4 sporadic cases (207). The deletion, flanked by direct repeats, removes 3 exons of the STX16 gene, which encodes syntaxin-16. Two other deletions within STX16 and larger deletions spanning both STX16 and its telomeric neighbor NPEPL1 have been identified in AD-PHP1B kindreds (208-211). In all these cases, maternal, but not paternal, inheritance of the STX16 deletion led to PTH resistance. Because STX16 is apparently not imprinted (208), loss of one copy of this gene is not predicted to underlie the PHP1B pathogenesis. Interestingly, two large deletions ablating NESP55 without any overlap with STX16 as the cause of PHP1B in families in whom affected individuals showed isolated loss of A/B methylation (211, 212). Note that the NESP55 region showed an apparent gain of methylation due to the deletion of the maternal allele.

 

In two other PHP1B kindreds, nearly identical deletions of the NESP55 DMR including exons 3 and 4 of the antisense transcript segregated with the disease (213). In this instance, however, the A/B DMR was not the only region to lose the differential methylation required to allow maternal expression of Gsα in the kidney. Maternal methylation was also lost in the regions of the XLαs and GNAS-AS1 promoters. Another kindred with these widespread epigenetic defects of GNAShas been described (214). The affected individuals in this kindred carried a maternally inherited deletion that removed antisense exons 3 and 4 with flanking intronic regions but not the NESP55 exon. Additional genomic deletions or rearrangements in the chromosomal regions comprising GNAS have also been identified and proposed to underlie the GNAS methylation abnormalities in some AD-PHP-Ib cases (215-219).

 

These PHP1B deletions point to two different imprinting control regions (ICRs) for the GNAS complex locus: one within the STX16 gene and the other at the NESP55 DMR. The ICR defined by the deletion at the neighboring STX16 gene seems to be in a different location in the mouse, because the targeted ablation of the region homologous to the 3-kb deletion caused neither Gnas methylation defects nor PTH resistance in mice (220). Recently, genome-wide methylation analysis of embryonic stem cells indicated that the A/B region is modestly hypomethylated compared to differentiated cells (221, 222), suggesting that this imprinted region differs from most other imprinted loci and is regulated critically in the early embryo. Subsequently, a study showed that deleting either the maternal STX16-ICR or the maternal NESP55-ICR results in significant further A/B hypomethylation in human embryonic stem cells (hESCs) (223). Moreover, while wild-type hESCs recovered their methylation following a transient inhibition of the maintenance DNA methyltransferase DNMT1 (mimicking the global demethylation process in the preimplantation embryo), the cells with maternally deleted STX16- or NESP55-ICR failed to regain methylation (223). This study also showed that the shortest region of overlap among the PHP1B-causing STX16 deletions was shown to harbor a pluripotent cell-specific enhancer element for the NESP55 promoter on the maternal allele (223). Taken together with a mouse study implicating NESP55 transcription in the regulation of maternal GNAS imprints (224), these findings strongly suggest that the GNAS exon A/B imprint is controlled, at least partly, in the early embryo by the NESP55 transcript that relies on the long-range enhancer within STX16. Thus, perturbation of this mechanism appears to be the underlying cause of the GNAS methylation defects observed in familial PHP1B cases.

 

Sporadic PHP1B cases also show broad GNAS epigenetic defects that involve A/B. In some of these cases, paternal uniparental disomy of different chromosome 20 segments have been reported as the likely cause of PHP1B in several such cases (225-229). The cause of the epigenetic defects and PTH resistance, however, remains unknown for most cases of sporadic PHP1B. GNAS methylation defects have been identified in some cases with hypomethylation at multiple maternally methylated imprinted regions (230-233). In fact, some of those cases show both PTH resistance and the clinical features resulting from the methylation changes of the other loci, such as Beckwith-Wiedemann Syndrome.

 

A recent study revealed that, in addition to the exon A/B DMR, methylation at a new GNAS region close to the GNAS-AS1 promoter (termed GNAS-AS2), is lost in patients who carry STX16 deletions (234). Note that this region is also affected in those cases that display broad GNAS methylation changes. Recently, two distinct subdomains with the GNAS-AS2 region have been identified, and a patient with partial loss of A/B methylation showed gain-of-methylation in one subdomain and no alteration in the other (235). The effect of methylation changes at GNAS-AS2 has yet to be determined at the level of gene expression, and their pathophysiologic significance is unclear. Two distinct PHP1B families have been recently described to carry maternal retrotransposon insertions in the large intron between exon XL and A/B of the maternal GNAS allele (236, 237). These cases had apparently normal levels of GNAS-AS2 methylation (235, 237), reflecting, perhaps, that the deleterious genetic alteration is located downstream of this DMR. The mechanism by which these retrotransposons cause A/B hypomethylation may entail perturbation of NESP55 transcription. The inserted sequence comprises multiple polyadenylation signals (AAUAAA), which may truncate the transcript prematurely, and one of the studies showed that the level of NESP55 transcript was reduced in patient-derived induced pluripotent cells (236).

 

A study compared the clinical phenotypes of PHP1B patients who show isolated A/B loss of methylation to those with broad GNAS methylation defects (238). No clinical differences could be established according to the pattern of GNASepigenetic defects, although serum PTH levels were significantly higher in females with broad GNAS methylation defects than females with isolated loss of 1A methylation. Another study also found an intrauterine growth advantage for both AD-PHP-1b and sporadic PHP-1b cases, but the results indicate that the sporadic cases are not as markedly growth accelerated as AD-PHP-1b cases at birth (239).

 

Contrary to the classical understanding that AHO features are unique to PHP1A, some studies have identified patients with PTH resistance and AHO features who show GNAS epigenetic defects rather than Gsα coding mutations (200, 240-242). Thus, there may be some overlap between the clinical and molecular features of PHP1A and PHP1B. It is possible that the AHO features observed in patients with GNAS epigenetic defects result from a genetic mechanism that is similar to the mechanism underlying the hormone resistance in PHP1A patients, i.e., due to monoallelic Gsα expression in additional tissues.

 

A PHP1B family with a novel Gsα mutation, deletion of isoleucine-382 in the carboxyl terminus has been described (243). In transfected cells this mutation led to uncoupling from the PTHR1 and isolated PTH resistance but not from other receptors, including TSH receptor. However, the same mutation showed uncoupling from multiple receptors, questioning the role of this mutation in the pathogenesis of PHP1B in this family. Such mutations within Gsα coding exons are likely to be a rare cause of PHP1B (166).  

 

PHP1c and PHP2

 

Patients with PHP1c have multiple hormone resistance but normal Gsα activity. The defect may be in other components of the receptor-adenylate cyclase system, such as the catalytic unit, but some PHP1c cases have been reported to carry Gsα coding mutations (244). These mutations render the Gsα protein unable to mediate cAMP generation in response to receptor activation but do not affect basal adenylate cyclase stimulating activity or the ability to be activated by non-hydrolyzable GTP analogs (244-246). Thus, some forms of PHP1c appear to be an allelic variant of PHP1A. Finally, patients with PHP2 have a normal urinary cAMP response to PTH but an impaired phosphaturic response (247). The defect could be in the cAMP-dependent protein kinase (PKA), one of its substrates or targets, or in a component of the PTH-PKC signaling pathway.

 

Impaired PTH-induced phosphaturia with normal nephrogenous cAMP formation (i.e., PHP2) appears as the least common form of PHP.  PHP2 is a sporadic disorder, but a familial form of PHP2 has been reported (248). In addition, a self-limited form of this disease in newborns has also been described, suggesting that it is transient in nature (249-251). The etiology and pathophysiological mechanisms behind this PHP variant remain unknown.  Because patients show adequate nephrogenous cAMP generation in response to exogenous PTH, molecular defects downstream of cAMP production are implicated, such as protein kinase A (247).  Accordingly, a study (252) has discovered a heterozygous mutation of the gene encoding the regulatory subunit of PKA (PRKAR1A) in three patients with multiple hormone resistance and acrodysostosis, a form of skeletal dysplasia that includes severe brachydactyly type-E and other skeletal findings that resemble AHO (also known as Maroteaux-Malamut syndrome (253, 254).  Several other variants of PRKAR1A have also been identified in other patients with a similar phenotype (255-258). These mutations, including the recurrent mutation R368X leading to the truncation of the C-terminal 14 residues, impair cAMP binding to the regulatory subunit, thereby blocking the activation of PKA (252, 259-261). In addition to acrodysostosis, patients carrying this mutation display evidence for target organ resistance to PTH, thyrotropin, growth hormone-releasing hormone, and gonadotropins, but these findings are accompanied by elevated basal plasma and urinary cAMP levels and with an apparently normal cAMP response to exogenous PTH administration. In certain other patients with acrodysostosis, but mostly without hormone resistance, it has been shown that the disease is caused by missense mutations in PDE4D, which encodes a cAMP phosphodiesterase (258, 262, 263). Given that PDE4D is an enzyme that reduces the intracellular cAMP concentration, the PDE4D mutations are likely to be gain-of-function (264).  The type of acrodysostosis caused by PRKAR1A mutations has been termed acrodysostosis-1 (MIM#101800), while the one caused by PDE4D mutations acrodysostosis-2 (MIM#614613). In addition, another subtype of cAMP phosphodiesterase, PDE3A, is affected in another disorder characterized by severe hypertension and brachydactyly type-E with short stature (154, 265), underscoring the importance of cAMP signaling in skeletal development and the regulation of vascular tone.

 

Other Phenotypes Associated with GNAS Mutations

 

In contrast to the PHP phenotype associated with inactivating GNAS mutations, a different form of sporadic bone disease, (polyostotic fibrous dysplasia) results from de novo GNAS mutations that cause constitutive Gsα activity (266). A more severe form of this disease (panostotic fibrous dysplasia) with hyperphosphatasia and hyperphosphaturic rickets has also been described (267, 268) . Patients carrying these activating mutations are mosaic for mutant and wild-type cells, indicating that the mutation is acquired during postzygotic development. These mutations affect the arginine residue at position 201 (exon 8) and, rarely, the glutamine at 227 (exon 9), and inhibit the intrinsic GTP hydrolase activity of Gsα, thereby leading to constitutive activity. Such constitutively activating mutations of GNAS are also found in a variety of endocrine and non-endocrine tumors, such as growth hormone-secreting adenomas (269) . A missense mutation in exon 13 (A366S) results in a Gsα protein that is unstable at 37°C, but constitutively active at lower temperatures (270, 271). Affected patients have PHP due to PTH resistance and precocious puberty (testotoxicosis) due to hormone-independent constitutive activation of luteinizing hormone receptors at lower ambient temperatures in the testes. Another Gsα mutant carrying Ala-Val-Asp-Thr amino acid repeats in the guanine-binding domain has been described in a patient with neonatal diarrhea and PTH resistance (272). In this instance, the mutant protein is unstable and localized to the cytoplasm rather than plasma membrane, which explains the hormone resistance. On the other hand, this mutation increases the rate of GDP-GTP exchange and, thus, confers overactivity. The increased activity of Gsα seems to be evident during the neonatal period in the gut, where the mutant localizes to the plasma membrane, thus explaining the diarrhea phenotype. Additional cases with missense Gsα mutations have been reported, presenting with clinical findings that likely reflect both gain and loss of Gsα function (273, 274).

 

Inactivating GNAS mutations have also been identified in patients with congenital osteoma cutis and progressive osseous heteroplasia (POH), suggesting that these connective tissue conditions are another variant in the phenotypic spectrum of GNAS-related disease (275-278). No genotype-phenotype correlation has been revealed regarding these disorders, as the same mutation can be associated with either typical AHO features or severe ossifications that involve deep connective tissues and skeletal muscle (279). Nonetheless, patients with POH inherit the GNAS mutation from their fathers or acquire this mutation de novo on the paternal GNAS allele. This parent-of-origin specific inheritance of POH was established by analyzing 18 unrelated kindreds with this disorder (280). In a single, three generation, kindred, the inheritance of the mutation from males led to POH, while the inheritance of the same mutation from females led to typical AHO. It thus appears likely that alterations in the activity of a paternally expressed GNAS product, such as XLαs, contribute to the pathogenesis of POH. However, POH-like features have also been seen in some patients with maternally inherited GNAS mutations (281). A study revealed that the distribution of POH lesions follows dermomyotomes and shows a tendency for one-sidedness, suggesting that post-zygotic second hits may contribute to the development of these lesions on top of the inherited heterozygous mutations of GNAS (282).

 

Differential Diagnosis and Genetic Counseling

 

Patients with dysmorphic features resembling AHO may require careful endocrinologic work-up to confirm and delineate the form of PHP that is present. Similar studies of family members may also be warranted, since the biochemical and clinical features vary within families. If PHP1A with AHO is established, genetic counseling may aid in understanding the multisystemic nature of the disorder, particularly in relation to the patient's growth and development, and later-onset connective tissue complications. For either PHP1A or PHP1B, extensive counseling may be required to adequately explain the various implications of paternal imprinting for the parent-specific recurrence risks in offspring. Germline mosaicism has been reported (176) , which is clearly important in assessing risks for recurrence in future sibs of a singleton family. Given the recently described complexities in the molecular, biochemical, and physical features of PHP1A and PHP1B, molecular testing is critical for achieving a clear diagnosis and validating the inheritance pattern in any given family.

 

THE PARATHYROID HORMONE RECEPTOR AND SKELETAL DYSPLASIAS

 

PTHR1 is a family B G protein-coupled receptor that signals through multiple different G proteins including Gsα (283). It responds to two ligands, PTH and the PTH-related peptide (PTHrP). It would thus be predicted that deleterious mutations might show resistance to PTH, as well as evidence for a defect of PTHrP action. Functional polymorphisms in the PTHR1 are associated with adult height and bone mineral density (284), emphasizing the role that the receptor and its ligands play in endochondral bone formation. Inactivating or loss-of-function mutations in the PTHR1 have been implicated in the molecular pathogenesis of Blomstrand lethal chondrodysplasia (BLC; MIM#215045), and other skeletal dysplasias and dental abnormalities (285). The rare, recessive BLC is characterized by short-limbed dwarfism with craniofacial malformations, hydrops, hypoplastic lungs and aortic coarctation (286-290). The bones show accelerated endochondral ossification and deficient remodeling. The Blomstrand disease has been subdivided into type I, which refers to the severe (classical) form, and type II, which refers to a relatively milder variant, and the difference between severity is attributed to complete or incomplete inactivation of the PTHR1, respectively (291, 292). A milder form of recessively inherited skeletal dysplasia, known as Eiken syndrome (MIM#600002), has also been linked to mutations of PTHR1 (293). Dominantly acting PTHR1 mutations have been identified in endochondromas of patients with enchondromatosis (Ollier's disease - MIM#166000), a familial disorder with evidence of autosomal dominance characterized by multiple benign cartilage tumors, and a predisposition to malignant osteocarcinomas (294, 295). As many patients with Ollier’s disease do not have PTHR1 mutations, it is likely that the condition is genetically heterogeneous (296). Dominantly inherited symmetrical enchondromatosis is associated with duplication of 12p11.23 to 12p11.22 that includes the PTHLH gene encoding PTHrP suggesting that abnormal PTHR1 signaling may underlie this unusual form of endochondromatosis (297). In addition, some cases of autosomal dominant nonsyndromic primary failure of tooth eruption (PFE) are due to loss-of-function mutations in the PTHR1 that are dominantly acting, leading to haploinsufficiency of the receptor (298-302) .

 

HYPOMAGNESEMIA

 

In humans, hypomagnesemia leads to a suppression of parathyroid hormone release and some degree of peripheral resistance. Although the exact molecular mechanism underlying the suppression of the parathyroid gland in hypomagnesemia is unknown, it is important to recognize that laboratory testing in cases of hypocalcemia with reduced PTH should include measurement of serum magnesium, particularly in newborns (303). Primary hypomagnesemia with secondary hypocalcemia (HSH) is an autosomal recessive disorder characterized by neuromuscular symptoms in infancy due to extremely low levels of serum magnesium and moderate to severe hypocalcemia. Homozygous mutations in the magnesium transporter gene transient receptor potential cation channel member 6 (TRPM6) cause the disease. HSH, a potentially lethal condition, can be misdiagnosed as primary hypoparathyroidism (304). Long-term prognosis after treatment with high dose of oral magnesium supplementation is good. Hypomagnesemia is also associated with long-term use of proton-pump inhibitors that decrease the luminal pH of the intestine by acting on the enterocyte apical TRPM6/7 channels (305, 306).

 

MANAGEMENT OF HYPOPARATHYROIDISM

 

Calcium and Vitamin D.

 

The goal of treatment in hypoparathyroid states is to raise the serum calcium sufficiently to alleviate acute symptoms of hypocalcemia and prevent the chronic complications (307, 308). The calcium concentration required for this purpose is generally in the low-normal range. It is equally important to ensure that treatment does not result in hypercalcemia, as even transient hypercalcemia could lead to nephrocalcinosis and renal failure.  Acute or severe symptomatic hypocalcemia is best treated with intravenous calcium infusion. Initial doses of 2 to 5 millimoles of elemental calcium as the gluconate salt can be given over a 10 to 20 minute period, followed by 2 millimoles elemental calcium per hour as a maintenance dose, to be adjusted according to symptoms and biochemical response. Care must be taken to ensure that the infusion does not extravasate, as this can lead to severe tissue damage. Where possible treatment through central access is preferred. Ionized or total calcium levels should be monitored frequently. Doses in children 5 to 14 years of age need to be adjusted for body weight, while neonates and infants require age-specific dosing. If present, hyperphosphatemia, alkalosis and hypomagnesemia should be corrected concomitantly. Post-surgical hypocalcemia after thyroid or parathyroid surgery is now rarely severe and usually transient with appropriate management (309). However, the occasional patient can represent a significant problem, particularly if the indication for surgery is chronic hyperparathyroidism, and the post-operative hypoparathyroid state is permanent (310). The long-term effects of standard therapy, hypercalciuria, nephrolithiasis, nephrocalcinosis, ectopic tissue calcification and mood changes, remain a concern (311).

 

The mainstay of chronic treatment is oral calcium and activated vitamin D (calcitriol), which should be started as soon as possible to allow reduction and discontinuation of the intravenous calcium. Oral calcium comes in several forms, but calcium carbonate is generally the least expensive. A total of 20 to 80 millimoles elemental calcium daily (2 to 8 g calcium carbonate per day) is generally effective, but should be given in divided doses and adjusted on the basis of gastro-intestinal tolerance, relief of hypocalcemic symptoms, and appropriate biochemical response. Vitamin D is preferably administered as calcitriol (0.25 to 1.0 micrograms per day), but, with some conditions, pharmacological doses of cholecalciferol or ergocalciferol or calcidiol may be less expensive and equally efficacious (312). Cholecalciferol and ergocalciferol doses are more difficult to titrate, and given their long duration of action, any overdoses can result in sustained toxicity. It is, therefore, appropriate to institute a starting dose of 25,000 IU/day and titrate upwards (to 100,000 IU/daily) with an assessment of serum and urinary parameters afterward with follow-up at 6 and 12 months, even if the patient is relatively asymptomatic. However, the use of active vitamin D (calcitriol or alphacalcidol) is recommended given that the lack of PTH along with the accompanying hyperphosphatemia reduces renal conversion of 25-hydroxyvitamin D to active vitamin D (307, 308). Serum calcium and 24-hour urinary excretion should be carefully monitored when therapy is started and continued until the dosing is stabilized. Hypercalciuria that occurs as treatment is initiated, even prior to the normalization of the serum calcium, may warrant an assessment of nephrocalcinosis by renal ultrasound. Consequently, only a low-normal serum calcium concentration may be attainable, but many patients feel well enough that there is no need to entirely normalize the serum calcium. In this way, the risk of renal failure due to chronic hypercalciuria − especially problematic in patients with CASR activating mutations (6, 7) − is minimized. Even after normalization or near-normalization of serum calcium, a significant number of patients report problems with fatigue, exhaustion, and mood disturbances (e.g., depression, anxiety, hostility, and paranoid ideation) not in keeping with the degree of hypocalcemia, suggesting that there may be non-calcitropic effects of PTH not remedied by maintenance of normocalcemia alone (311). In an epidemiological and health-related quality of life study from Norway, postsurgical hypoparathyroid patients scored worse than those with nonsurgical hypoparathyroidism or pseudohypoparathyroidism (313), providing further support for the notion of direct effects of PTH on mood.

 

In pseudohypoparathyroidism, calcitriol (and not other forms of vitamin D) should be used for the treatment, because the PTH resistance in the proximal tubule does not allow for the efficient synthesis of 1,25(OH)2D from 25-hydroxyvitamin D.  In pseudohypoparathyroidism type 1A, there is also a degree of PTH resistance in the bone due to haploinsufficiency of Gsα.  However, in pseudohypothyroidism type 1B, the bone is fully sensitive to PTH, so monitoring serum PTH levels during treatment is critical with the aim of normalizing or reducing PTH levels as much as possible (314, 315). Hypercalciuria as a result of the calcitriol and calcium treatment is a lesser concern in pseudohypoparathyroidism because PTH actions in the distal tubule are still functional, preventing excess loss of calcium in the urine and providing greater protection against nephrocalcinosis.

 

Hormone Replacement Therapy  

 
Hormone replacement has been advocated as a potentially superior form of treatment for decades but only recently have preparations of recombinant human hormone –– teriparatide (PTH 1-34), full-length parathyroid hormone (PTH 1-84), and abaloparatide (PTHrP analog) — become available. In 2015, the U.S. Food and Drug Administration (FDA) approved recombinant human (rh) PTH (1-84) for the management of hypoparathyroidism (316). This provided an additional therapeutic option for the management of those patients who demonstrate poor control with the standard calcium and active vitamin D supplemental therapy. The FDA indication was for subjects with hypoparathyroidism of any etiology, except ADH, but including postsurgical cases. The FDA did not limit the duration of its use but approved rhPTH(1-84) with a “black box” warning because of the history of rat osteosarcoma and PTH use (317). However, no evidence for this in primates or in clinical use has been forthcoming (318), and the ‘black box’ warning has since been withdrawn (319).

 

The use of PTH in hypoparathyroidism was demonstrated initially with the amino-terminal fragment of PTH, teriparatide [PTH(1–34)] (320). Beneficial control in children and in adults occurred when teriparatide was administered daily, with better control when the peptide was administered in twice-daily dosing regimens (320-324). With a pump delivery system by which teriparatide could be administered continuously (325, 326), urinary calcium excretion fell, and markers of bone turnover normalized. A smaller daily dose was required with pump delivery vs multiple daily dosing regimens. An open-label trial of PTH(1–34) in adult subjects with postsurgical hypoparathyroidism showed improvement in quality of life (327). Beneficial effects on calcium homeostasis have also been demonstrated in specific ADH cases with activating CaSR mutations (328, 329).

 

The full-length PTH (1-84) mimics the secreted product of the parathyroid gland, and its longer biological half-life (than PTH(1-34) makes once-daily dosing feasible in the treatment of hypoparathyroidism (330-332). Studies by several groups have noted a substantial reduction in the requirement for calcium and active vitamin D (333-335); only transient reductions in urinary calcium excretion (331); a tendency for lumbar spine bone mineral density (BMD) to increase and that of the distal one-third radius to fall (334); a rapid increase in bone turnover, assessed by circulating markers and dynamic histomorphometric analyses of bone that achieves a new steady state that is higher than baseline values within 2–3 years (336); and improvements in quality of life in some studies (333, 337).

 

In a placebo-controlled 24-week clinical trial of rhPTH (1-84) in 130 hypoparathyroid patients the primary endpoints of a reduction by 50% in calcium supplements and in active vitamin D along with maintenance of the serum calcium were met in over half of the study subjects (338). There was a greater percentage of subjects in whom active vitamin D could be eliminated entirely while taking no more than 500 mg of oral calcium daily. The drug was titrated from 50 to 100 μg/d, with just over half of the subjects needing the highest dose. The rhPTH(1-84) reduced serum phosphate levels, improved the calcium-phosphate product, and maintained 1,25(OH)2D and serum calcium levels in the normal range (339). In addition, therapy with a long-acting prodrug of PTH(1-34), TransCon PTH (palopegteriparatide), in hypoparathyroidism has been shown to improve scores in quality-of-life measures (340). However, despite these early positive results, the inconvenient route of administration, daily or twice daily subcutaneous injection, leads to most patients opting for conventional treatment with oral calcium and calcitriol.

 

The manufacturer of rhPTH(1-34) has recently decided to discontinue this product at the end of 2024 due to an unresolved supply issue (https://www.takeda.com/newsroom/statements/2022/discontinue-manufacturing-natpar-natpara/). In addition, the use of teriparatide or aboloparatide for hypoparathyroidism has not been approved by the FDA. Therefore, no available FDA-approved hormone replacement therapies currently exist for the management of this disorder.  

  

Calcilytics  

 

Calcilytics are small molecule allosteric modulators of the CASR that antagonize the calcium-sensing receptor and promote PTH secretion and are a promising alternative for disorders with intact but hypofunctioning parathyroid glands (341). Calcilytics inhibit the activation of the CASR in both the parathyroid and renal tubule, and thus, they not only promote PTH secretion but also increase renal calcium reabsorption and are, therefore, of potential interest for the treatment of ADH1. In contrast, clinical studies in patients with ADH1 treated with PTH(1-34) led to better control of blood calcium levels (324), but the effects of the activated CASR in the kidney led to continued increases in urinary calcium excretion, different from patients with postsurgical hypoparathyroidism (326, 329). Thus, while FDA approval was given for PTH treatment of hypoparathyroidism, ADH1 was excluded from the indication.

 

In cell culture experiments studying activating CASR mutants, calcilytics normalize the left-ward shift of the calcium response curve (342, 343). The utility of calcilytics was further demonstrated in studies of mice harboring activating Casrmutations. In one study, two knock-in mouse models of ADH1 with activating mutations in the Casr were generated. Daily oral administration of the calcilytic JTT-305/MK-3442 to these mice increased serum PTH and calcium levels and reduced urinary calcium excretion (310). Intraperitoneal injection of the calcilytic NPS2143 in the nuf mouse model of ADH1, transiently increased circulating PTH and calcium levels without increasing urinary calcium levels (342). In a preliminary clinical study, IV administration of the calcilytic NPSP795 to five patients with ADH1 increased their plasma PTH levels and decreased their fractional urinary calcium excretion (344). Calcilytics comprise two main classes of compounds; the amino alcohols (e.g., NPS2143, NPSP795, JTT-305/MK-5442) and the quinazolinones (e.g., ATF936 and AXT914) (341). While both classes of compounds corrected the gain-of-function properties of several of the ADH1 CASR mutations tested in vitro, a subset of mutations involving NPS2143 binding sites within the transmembrane domain of the CASR are not fully corrected with NPS2143 but are normalized with the quinazolinone drugs (ATF936 and AXT914) (345-347). Whether this is reflected in mouse model studies and clinical situations remains to be determined.

 

Cases of hypoparathyroidism presenting as ADH but without CASR mutations have been found to have activating mutations of the gene encoding Gα11, the alpha-subunit of the heterotrimeric G protein that couples the CASR to signaling pathways (348, 349). The syndrome has been designated ADH2. Even though Gα11 is downstream of the CASR, in vitro studies showed that the calcilytic NPS2143 rectifies the altered Ca2+ signaling of the overactive mutants (350). Knock-in mice harboring an ADH2 Gα11 activating mutation faithfully replicate ADH2 (351). Treatment with the calcilytic NPS2143 or a Gα11/q-specific inhibitor, YM-254890 (352), increased circulating PTH and calcium levels in the heterozygous mutant mice (351). Thus, calcilytics, by blocking the renal CASR, may have potential use for treating ADH1 and ADH2, as well as other forms of hypoparathyroidism.

 

Other Therapies  

 

If the serum calcium attainable with oral calcium and calcitriol is below the normal range and the patient remains symptomatic, then a trial of a thiazide diuretic may be considered, with the aim of reducing the hypercalciuria to raise the serum calcium further. The argument for efficacy seems greatest for responsive forms of autosomal dominant hypocalcemia due to activating CaSR mutations, since the thiazide-sensitive transporter, SLC12A3 (MIM#600968), is a downstream target of and is suppressed by activated CaSR in the kidney. For reasons that are not clear, however, thiazides work well in only a subset of patients (353). It is critical to monitor serum potassium and magnesium levels, as thiazide use can increase renal losses of these cations with resulting hypokalemia and hypomagnesemia. Some authorities suggest thiazides should not be used in APS1 patients with adrenal insufficiency and in ADH1 patients with Bartter syndrome type V (307, 308).

 

As the serum calcium is normalized, elevated serum phosphate concentrations generally decline, but phosphate-binding gels such as aluminum hydroxide are occasionally helpful in reducing hyperphosphatemia at the beginning of therapy or in cases where there is persistent hyperphosphatemia. Patients who develop intracranial calcifications may experience seizures related to chronic neuropathic changes, and it may be necessary to add appropriate anti-epileptic medication(s). In all chronically hypocalcemic patients, ocular assessments should be performed periodically.

 

In cases with documented abnormalities of the somatotropin axis, the growth hormone replacement therapy is effective but has to be initiated as soon as possible (315, 354, 355).

 

REFERENCES

 

  1. Shoback DM, Bilezikian JP, Costa AG, Dempster D, Dralle H, Khan AA, et al. Presentation of Hypoparathyroidism: Etiologies and Clinical Features. The Journal of Clinical Endocrinology &amp; Metabolism. 2016;101(6):2300-12.
  2. Clarke BL, Brown EM, Collins MT, Jüppner H, Lakatos P, Levine MA, et al. Epidemiology and Diagnosis of Hypoparathyroidism. The Journal of clinical endocrinology and metabolism. 2016;101(6):2284-99.
  3. Schafer AL, and Shoback DM. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al. eds. Endotext. South Dartmouth (MA); 2000.
  4. Fisher NG, Armitage A, McGonigle RJ, and Gilbert TJ. Hypocalcaemic Cardiomyopathy; the relationship between myocardial damage, left ventricular function, calcium and ECG changes in a patient with idiopathic hypocalcaemia. European Journal of Heart Failure. 2001;3(3):373-6.
  5. Tziomalos K, Kakavas N, Kountana E, Harsoulis F, and Basayannis E. Reversible dilated hypocalcaemic cardiomyopathy in a patient with primary hypoparathyroidism. Clinical endocrinology. 2006;64(6):717-8.
  6. Lienhardt A, Bai M, Lagarde J-P, Rigaud M, Zhang Z, Jiang Y, et al. Activating Mutations of the Calcium-Sensing Receptor: Management of Hypocalcemia. The Journal of Clinical Endocrinology &amp; Metabolism. 2001;86(11):5313-23.
  7. Obermannova B, Sumnik Z, Dusatkova P, Cinek O, Grant M, Lebl J, et al. Novel calcium-sensing receptor cytoplasmic tail deletion mutation causing autosomal dominant hypocalcemia: molecular and clinical study. European Journal of Endocrinology. 2016;174(4):K1-K11.
  8. McKusick VA. Mendelian Inheritance in Man and its online version, OMIM. Am J Hum Genet. 2007;80(4):588-604.
  9. Arnold A, Horst SA, Gardella TJ, Baba H, Levine MA, and Kronenberg HM. Mutation of the signal peptide-encoding region of the preproparathyroid hormone gene in familial isolated hypoparathyroidism. J Clin Invest. 1990;86:1084-7.
  10. Datta R, Waheed A, Shah GN, and Sly WS. Signal sequence mutation in autosomal dominant form of hypoparathyroidism induces apoptosis that is corrected by a chemical chaperone. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(50):19989-94.
  11. Sunthornthepvarakul T, Churesigaew S, and Ngowngarmratana S. A Novel Mutation of the Signal Peptide of the Preproparathyroid Hormone Gene Associated with Autosomal Recessive Familial Isolated Hypoparathyroidism*. The Journal of Clinical Endocrinology &amp; Metabolism. 1999;84(10):3792-6.
  12. Ertl D-A, Stary S, Streubel B, Raimann A, and Haeusler G. A novel homozygous mutation in the parathyroid hormone gene (PTH) in a girl with isolated hypoparathyroidism. Bone. 2012;51(3):629-32.
  13. Lee S, Mannstadt M, Guo J, Kim SM, Yi H-S, Khatri A, et al. A Homozygous [Cys25]PTH(1-84) Mutation That Impairs PTH/PTHrP Receptor Activation Defines a Novel Form of Hypoparathyroidism. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2015;30(10):1803-13.
  14. Parkinson DB, and Thakker RV. A donor splice site mutation in the parathyroid hormone gene is associated with autosomal recessive hypoparathyroidism. Nature Genetics. 1992;1(2):149-52.
  15. Grigorieva IV, and Thakker RV. Transcription factors in parathyroid development: lessons from hypoparathyroid disorders. Annals of the New York Academy of Sciences. 2011;1237(1):24-38.
  16. Bowl MR, Nesbit MA, Harding B, Levy E, Jefferson A, Volpi E, et al. An interstitial deletion-insertion involving chromosomes 2p25.3 and Xq27.1, near SOX3, causes X-linked recessive hypoparathyroidism. The Journal of clinical investigation. 2005;115(10):2822-31.
  17. Pollak MR, Brown EM, Estep HL, McLaine PN, Kifor O, Park J, et al. Autosomal dominant hypocalcaemia caused by a Ca2+-sensing receptor gene mutation. Nature Genetics. 1994;8:303-7.
  18. Hendy GN, D'Souza-Li L, Yang B, Canaff L, and Cole DEC. Mutations of the calcium-sensing receptor (CASR) in familial hypocalciuric hypercalcemia, neonatal severe hyperparathyroidism, and autosomal dominant hypocalcemia. Human Mutation. 2000;16(4):281-96.
  19. Cole DEC, Yun FHJ, Wong BYL, Shuen AY, Booth RA, Scillitani A, et al. Calcium-sensing receptor mutations and denaturing high performance liquid chromatography. Journal of Molecular Endocrinology. 2009;42(4):331-9.
  20. Hendy GN, Guarnieri V, and Canaff L. Progress in Molecular Biology and Translational Science. Elsevier; 2009:31-95.
  21. Hendy GN, Minutti C, Canaff L, Pidasheva S, Yang B, Nouhi Z, et al. Recurrent Familial Hypocalcemia Due to Germline Mosaicism for an Activating Mutation of the Calcium-Sensing Receptor Gene. The Journal of Clinical Endocrinology &amp; Metabolism. 2003;88(8):3674-81.
  22. Ray K, Hauschild BC, Steinbach PJ, Goldsmith PK, Hauache O, and Spiegel AM. Identification of the Cysteine Residues in the Amino-terminal Extracellular Domain of the Human Ca2+ Receptor Critical for Dimerization. Journal of Biological Chemistry. 1999;274(39):27642-50.
  23. Pidasheva S, D'Souza-Li L, Canaff L, Cole DEC, and Hendy GN. CASRdb: calcium-sensing receptor locus-specific database for mutations causing familial (benign) hypocalciuric hypercalcemia, neonatal severe hyperparathyroidism, and autosomal dominant hypocalcemia. Human Mutation. 2004;24(2):107-11.
  24. Hannan FM, Nesbit MA, Zhang C, Cranston T, Curley AJ, Harding B, et al. Identification of 70 calcium-sensing receptor mutations in hyper- and hypo-calcaemic patients: evidence for clustering of extracellular domain mutations at calcium-binding sites. Human Molecular Genetics. 2012;21(12):2768-78.
  25. Vargas-Poussou R, Huang C, Hulin P, Houillier P, Jeunemaitre X, Paillard M, et al. Functional Characterization of a Calcium-Sensing Receptor Mutation in Severe Autosomal Dominant Hypocalcemia with a Bartter-Like Syndrome. Journal of the American Society of Nephrology. 2002;13(9):2259-66.
  26. Watanabe S, Fukumoto S, Chang H, Takeuchi Y, Hasegawa Y, Okazaki R, et al. Association between activating mutations of calcium-sensing receptor and Bartter's syndrome. The Lancet. 2002;360(9334):692-4.
  27. Vezzoli G, Arcidiacono T, Paloschi V, Terranegra A, Biasion R, Weber G, et al. Autosomal dominant hypocalcemia with mild type 5 Bartter syndrome. J Nephrol. 2006;19(4):525-8.
  28. Hebert SC. Bartter syndrome. Current opinion in nephrology and hypertension. 2003;12(5):527-32.
  29. Carmosino M, Gerbino A, Hendy GN, Torretta S, Rizzo F, Debellis L, et al. NKCC2 activity is inhibited by the Bartter's syndrome type 5 gain-of-function CaR-A843E mutant in renal cells. Biol Cell. 2015;107(4):98-110.
  30. Nesbit MA, Hannan FM, Howles SA, Babinsky VN, Head RA, Cranston T, et al. Mutations affecting G-protein subunit alpha11 in hypercalcemia and hypocalcemia. N Engl J Med. 2013;368(26):2476-86.
  31. Mannstadt M, Harris M, Bravenboer B, Chitturi S, Dreijerink KM, Lambright DG, et al. Germline mutations affecting Galpha11 in hypoparathyroidism. N Engl J Med. 2013;368(26):2532-4.
  32. Li D, Opas EE, Tuluc F, Metzger DL, Hou C, Hakonarson H, et al. Autosomal dominant hypoparathyroidism caused by germline mutation in GNA11: phenotypic and molecular characterization. J Clin Endocrinol Metab. 2014;99(9):E1774-83.
  33. Piret SE, Gorvin CM, Pagnamenta AT, Howles SA, Cranston T, Rust N, et al. Identification of a G-Protein Subunit-α11 Gain-of-Function Mutation, Val340Met, in a Family With Autosomal Dominant Hypocalcemia Type 2 (ADH2). Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2016;31(6):1207-14.
  34. Mizuno N, and Itoh H. Functions and regulatory mechanisms of Gq-signaling pathways. Neurosignals. 2009;17(1):42-54.
  35. Hendy GN, and Cole DE. Ruling in a suspect: the role of AP2S1 mutations in familial hypocalciuric hypercalcemia type 3. J Clin Endocrinol Metab. 2013;98(12):4666-9.
  36. Lambert AS, Grybek V, Francou B, Esterle L, Bertrand G, Bouligand J, et al. Analysis of AP2S1, a calcium-sensing receptor regulator, in familial and sporadic isolated hypoparathyroidism. J Clin Endocrinol Metab. 2014;99(3):E469-73.
  37. Rogers A, Nesbit MA, Hannan FM, Howles SA, Gorvin CM, Cranston T, et al. Mutational analysis of the adaptor protein 2 sigma subunit (AP2S1) gene: search for autosomal dominant hypocalcemia type 3 (ADH3). J Clin Endocrinol Metab. 2014;99(7):E1300-5.
  38. Kanemura Y, Hiraga S, Arita N, Ohnishi T, Izumoto S, Mori K, et al. Isolation and expression analysis of a novel human homologue of the <i>Drosophila glial cells missing</i> (<i>gcm</i>) gene. FEBS Letters. 1999;442(2-3):151-6.
  39. Gunther T, Chen Z-F, Kim J, Priemel M, Rueger JM, Amling M, et al. Genetic ablation of parathyroid glands reveals another source of parathyroid hormone. Nature. 2000;406(6792):199-203.
  40. Okabe M, and Graham A. The origin of the parathyroid gland. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(51):17716-9.
  41. Ding C, Buckingham B, and Levine MA. Familial isolated hypoparathyroidism caused by a mutation in the gene for the transcription factor GCMB. Journal of Clinical Investigation. 2001;108(8):1215-20.
  42. Thomee C, Schubert SW, Parma J, Lê PQ, Hashemolhosseini S, Wegner M, et al. GCMB Mutation in Familial Isolated Hypoparathyroidism with Residual Secretion of Parathyroid Hormone. The Journal of Clinical Endocrinology & Metabolism. 2005;90(5):2487-92.
  43. Canaff L, Zhou X, Mosesova I, Cole DEC, and Hendy GN. Glial Cells Missing-2 (GCM2) transactivates the calcium-sensing receptor gene: effect of a dominant-negative GCM2 mutant associated with autosomal dominant hypoparathyroidism. Human Mutation. 2009;30(1):85-92.
  44. Bowl MR, Mirczuk SM, Grigorieva IV, Piret SE, Cranston T, Southam L, et al. Identification and characterization of novel parathyroid-specific transcription factor Glial Cells Missing Homolog B (GCMB) mutations in eight families with autosomal recessive hypoparathyroidism. Hum Mol Genet. 2010;19(10):2028-38.
  45. Mannstadt M, Bertrand G, Muresan M, Weryha G, Leheup B, Pulusani SR, et al. Dominant-negative GCMB mutations cause an autosomal dominant form of hypoparathyroidism. The Journal of clinical endocrinology and metabolism. 2008;93(9):3568-76.
  46. Mirczuk SM, Bowl MR, Nesbit MA, Cranston T, Fratter C, Allgrove J, et al. A Missense<i>Glial Cells Missing Homolog B</i>(<i>GCMB</i>) Mutation, Asn502His, Causes Autosomal Dominant Hypoparathyroidism. The Journal of Clinical Endocrinology &amp; Metabolism. 2010;95(7):3512-6.
  47. Hendy GN, and Cole DEC. Hypoparathyroidism. Springer Milan; 2015:167-75.
  48. Maret A, Ding C, Kornfield SL, and Levine MA. Analysis of the<i>GCM2</i>Gene in Isolated Hypoparathyroidism: A Molecular and Biochemical Study. The Journal of Clinical Endocrinology &amp; Metabolism. 2008;93(4):1426-32.
  49. D'Agruma L, Coco M, Guarnieri V, Battista C, Canaff L, Salcuni AS, et al. Increased Prevalence of the<i>GCM2</i>Polymorphism, Y282D, in Primary Hyperparathyroidism: Analysis of Three Italian Cohorts. The Journal of Clinical Endocrinology &amp; Metabolism. 2014;99(12):E2794-E8.
  50. Guan B, Welch JM, Sapp JC, Ling H, Li Y, Johnston JJ, et al. GCM2-Activating Mutations in Familial Isolated Hyperparathyroidism. American journal of human genetics. 2016;99(5):1034-44.
  51. Lackey AE, and Muzio MR. StatPearls. Treasure Island (FL); 2023.
  52. McDonald-McGinn DM, Sullivan KE, Marino B, Philip N, Swillen A, Vorstman JAS, et al. 22q11.2 deletion syndrome. Nat Rev Dis Primers. 2015;1:15071-.
  53. Botto LD, May K, Fernhoff PM, Correa A, Coleman K, Rasmussen SA, et al. A Population-Based Study of the 22q11.2 Deletion: Phenotype, Incidence, and Contribution to Major Birth Defects in the Population. Pediatrics. 2003;112(1):101-7.
  54. Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, et al. Role of TBX1 in human del22q11.2 syndrome. The Lancet. 2003;362(9393):1366-73.
  55. Baldini A, Fulcoli FG, and Illingworth E. Current Topics in Developmental Biology. Elsevier; 2017:223-43.
  56. Scambler PJ. 22q11 Deletion Syndrome: A Role for TBX1 in Pharyngeal and Cardiovascular Development. Pediatric Cardiology. 2010;31(3):378-90.
  57. Guo C, Sun Y, Zhou B, Adam RM, Li X, Pu WT, et al. A Tbx1-Six1/Eya1-Fgf8 genetic pathway controls mammalian cardiovascular and craniofacial morphogenesis. The Journal of clinical investigation. 2011;121(4):1585-95.
  58. Huh S-H, and Ornitz DM. Beta-catenin deficiency causes DiGeorge syndrome-like phenotypes through regulation of Tbx1. Development (Cambridge, England). 2010;137(7):1137-47.
  59. Jerome LA, and Papaioannou VE. DiGeorge syndrome phenotype in mice mutant for the T-box gene, Tbx1. Nature Genetics. 2001;27(3):286-91.
  60. Ivins S, Lammerts van Beuren K, Roberts C, James C, Lindsay E, Baldini A, et al. Microarray analysis detects differentially expressed genes in the pharyngeal region of mice lacking Tbx1. Developmental Biology. 2005;285(2):554-69.
  61. Garg V, Yamagishi C, Hu T, Kathiriya IS, Yamagishi H, and Srivastava D. Tbx1, a DiGeorge Syndrome Candidate Gene, Is Regulated by Sonic Hedgehog during Pharyngeal Arch Development. Developmental Biology. 2001;235(1):62-73.
  62. Scire G, Dallapiccola B, Iannetti P, Bonaiuto F, Galasso C, Mingarelli R, et al. Hypoparathyroidism as the major manifestation in two patients with 22q11 deletions. American journal of medical genetics. 1994;52(4):478-82.
  63. Sykes K, Bachrach L, Siegel-Bartelt J, Ipp M, Kooh S, and Cytrynbaum C. Velocardiofacial syndrome presenting as hypocalcemia in early adolescence. Arch Pediatr Adolesc Med. 1997;151:745-7.
  64. Kapadia RK, and Bassett AS. Recognizing a common genetic syndrome: 22q11.2 deletion syndrome. CMAJ. 2008;178(4):391-3.
  65. Liu H, Abecasis GR, Heath SC, Knowles A, Demars S, Chen Y-J, et al. Genetic variation in the 22q11 locus and susceptibility to schizophrenia. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(26):16859-64.
  66. McDonald-McGinn DM, Hain HS, Emanuel BS, and Zackai EH. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, et al. eds. GeneReviews((R)). Seattle (WA); 1993.
  67. Burnside RD. 22q11.21 Deletion Syndromes: A Review of Proximal, Central, and Distal Deletions and Their Associated Features. Cytogenetic and Genome Research. 2015;146(2):89-99.
  68. Goodship J, Cross I, Scambler P, and Burn J. Monozygotic twins with chromosome 22q11 deletion and discordant phenotype. Journal of medical genetics. 1995;32(9):746-8.
  69. Hillebrand G, Siebert R, Simeoni E, and Santer R. DiGeorge syndrome with discordant phenotype in monozygotic twins. J Med Genet. 2000;37(9):E23-E.
  70. Miller JD, Bowker BM, Cole DE, and Guyda HJ. DiGeorge's syndrome in monozygotic twins. Treatment with calcitriol. Am J Dis Child. 1983;137(5):438-40.
  71. Alkalay AA, Guo T, Montagna C, Digilio MC, Dallapiccola B, Marino B, et al. Genetic dosage compensation in a family with velo-cardio-facial/DiGeorge/22q11.2 deletion syndrome. American journal of medical genetics Part A. 2011;155A(3):548-54.
  72. Carelle-Calmels N, Saugier-Veber P, Girard-Lemaire F, Rudolf G, Doray B, Guerin E, et al. Genetic Compensation in a Human Genomic Disorder. New England Journal of Medicine. 2009;360(12):1211-6.
  73. Jatana V, Gillis J, Webster BH, and Adès LC. Deletion 22q11.2 syndrome—Implications for the intensive care physician*. Pediatric Critical Care Medicine. 2007;8(5):459-63.
  74. Nagasaki K, Iwasaki Y, Ogawa Y, Kikuchi T, and Uchiyama M. Evaluation of parathyroid gland function using sodium bicarbonate infusion test for 22q11.2 deletion syndrome. Hormone research in paediatrics. 2011;75(1):14-8.
  75. Weinzimer SA. Endocrine aspects of the 22q11.2 deletion syndrome. Genetics in Medicine. 2001;3(1):19-22.
  76. Jensen TJ, Dzakula Z, Deciu C, van den Boom D, and Ehrich M. Detection of Microdeletion 22q11.2 in a Fetus by Next-Generation Sequencing of Maternal Plasma. Clinical Chemistry. 2012;58(7):1148-51.
  77. Tenhola S, Hendy GN, Valta H, Canaff L, Lee BSP, Wong BYL, et al. Cinacalcet Treatment in an Adolescent With Concurrent 22q11.2 Deletion Syndrome and Familial Hypocalciuric Hypercalcemia Type 3 Caused by<i>AP2S1</i>Mutation. The Journal of Clinical Endocrinology &amp; Metabolism. 2015;100(7):2515-8.
  78. Lima K, Abrahamsen TG, Wolff ABe, Husebye E, Alimohammadi M, Kämpe O, et al. Hypoparathyroidism and autoimmunity in the 22q11.2 deletion syndrome. European Journal of Endocrinology. 2011;165(2):345-52.
  79. Meek CL, Kaplan F, Pereira RS, and Viljoen A. Hypocalcemia following Treatment for Hyperthyroidism. Clinical Chemistry. 2011;57(6):811-4.
  80. Daw SCM, Taylor C, Kraman M, Call K, Mao J-i, Schuffenhauer S, et al. A common region of 10p deleted in DiGeorge and velocardiofacial syndromes. Nature Genetics. 1996;13(4):458-60.
  81. Gottlieb S, Driscoll DA, Punnett HH, Sellinger B, Emanuel BS, and Budarf ML. Characterization of 10p deletions suggests two nonoverlapping regions contribute to the DiGeorge syndrome phenotype. American journal of human genetics. 1998;62(2):495-8.
  82. Schuffenhauer S, Lichtner P, Peykar-Derakhshandeh P, Murken J, Haas OA, Back E, et al. Deletion mapping on chromosome 10p and definition of a critical region for the second DiGeorge syndrome locus (DGS2). European Journal of Human Genetics. 1998;6(3):213-25.
  83. Barakat AY, D'Albora JB, Martin MM, and Jose PA. Familial nephrosis, nerve deafness, andhypoparathyroidism. The Journal of Pediatrics. 1977;91(1):61-4.
  84. Bilous RW, Murty G, Parkinson DB, Thakker RV, Coulthard MG, Burn J, et al. Autosomal Dominant Familial Hypoparathyroidism, Sensorineural Deafness, and Renal Dysplasia. New England Journal of Medicine. 1992;327(15):1069-74.
  85. Lichtner P, Konig R, Hasegawa T, Van Esch H, Meitinger T, and Schuffenhauer S. An HDR (hypoparathyroidism, deafness, renal dysplasia) syndrome locus maps distal to the DiGeorge syndrome region on 10p13/14. Journal of medical genetics. 2000;37(1):33-7.
  86. Van Esch H, Groenen P, Daw S, Poffyn A, Holvoet M, Scambler P, et al. Partial DiGeorge syndrome in two patients with a 10p rearrangement. Clinical genetics. 1999;55(4):269-76.
  87. Van Esch H, Groenen P, Nesbit MA, Schuffenhauer S, Lichtner P, Vanderlinden G, et al. GATA3 haplo-insufficiency causes human HDR syndrome. Nature. 2000;406(6794):419-22.
  88. Ali A, Christie PT, Grigorieva IV, Harding B, Van Esch H, Ahmed SF, et al. Functional characterization of GATA3 mutations causing the hypoparathyroidism-deafness-renal (HDR) dysplasia syndrome: insight into mechanisms of DNA binding by the GATA3 transcription factor. Human Molecular Genetics. 2006;16(3):265-75.
  89. Nesbit MA. Hypoparathyroidism. Springer Milan; 2015:199-213.
  90. Nesbit MA, Bowl MR, Harding B, Ali A, Ayala A, Crowe C, et al. Characterization of GATA3 Mutations in the Hypoparathyroidism, Deafness, and Renal Dysplasia (HDR) Syndrome. Journal of Biological Chemistry. 2004;279(21):22624-34.
  91. Zahirieh A, Nesbit MA, Ali A, Wang K, He N, Stangou M, et al. Functional analysis of a novel GATA3 mutation in a family with the hypoparathyroidism, deafness, and renal dysplasia syndrome. J Clin Endocrinol Metab. 2005;90(4):2445-50.
  92. Grigorieva IV, Mirczuk S, Gaynor KU, Nesbit MA, Grigorieva EF, Wei Q, et al. Gata3-deficient mice develop parathyroid abnormalities due to dysregulation of the parathyroid-specific transcription factor Gcm2. The Journal of clinical investigation. 2010;120(6):2144-55.
  93. Kenny FM, and Linarelli L. Dwarfism and cortical thickening of tubular bones. Transient hypocalcemia in a mother and son. Am J Dis Child. 1966;111(2):201-7.
  94. Bergada I, Schiffrin A, Abu Srair H, Kaplan P, Dornan J, Goltzman D, et al. Kenny syndrome: description of additional abnormalities and molecular studies. Human Genetics. 1988;80(1):39-42.
  95. Caffey J. CONGENITAL STENOSIS OF MEDULLARY SPACES IN TUBULAR BONES AND CALVARIA IN TWO PROPORTIONATE DWARFS—MOTHER AND SON; COUPLED WITH TRANSITORY HYPOCALCEMIC TETANY. American Journal of Roentgenology. 1967;100(1):1-11.
  96. Hershkovitz E, and Parvari R. Hypoparathyroidism. Springer Milan; 2015:215-24.
  97. Abraham MB, Li D, Tang D, O'Connell SM, McKenzie F, Lim EM, et al. Short stature and hypoparathyroidism in a child with Kenny-Caffey syndrome type 2 due to a novel mutation in FAM111A gene. Int J Pediatr Endocrinol. 2017;2017:1.
  98. Isojima T, Doi K, Mitsui J, Oda Y, Tokuhiro E, Yasoda A, et al. A recurrent de novo <i>FAM111A</i> mutation causes kenny–caffey syndrome type 2. Journal of Bone and Mineral Research. 2014;29(4):992-8.
  99. Unger S, Gorna MW, Le Bechec A, Do Vale-Pereira S, Bedeschi MF, Geiberger S, et al. FAM111A mutations result in hypoparathyroidism and impaired skeletal development. American journal of human genetics. 2013;92(6):990-5.
  100. Alabert C, Bukowski-Wills J-C, Lee S-B, Kustatscher G, Nakamura K, de Lima Alves F, et al. Nascent chromatin capture proteomics determines chromatin dynamics during DNA replication and identifies unknown fork components. Nature cell biology. 2014;16(3):281-93.
  101. Teebi AS. Hypoparathyroidism, retarded growth and development, and dysmorphism or Sanjad-Sakati syndrome: an Arab disease reminiscent of Kenny-Caffey syndrome. J Med Genet. 2000;37(2):145.
  102. Parvari R, Hershkovitz E, Kanis A, Gorodischer R, Shalitin S, Sheffield VC, et al. Homozygosity and linkage-disequilibrium mapping of the syndrome of congenital hypoparathyroidism, growth and mental retardation, and dysmorphism to a 1-cM interval on chromosome 1q42-43. American journal of human genetics. 1998;63(1):163-9.
  103. Parvari R, Hershkovitz E, Grossman N, Gorodischer R, Loeys B, Zecic A, et al. Mutation of TBCE causes hypoparathyroidism-retardation-dysmorphism and autosomal recessive Kenny-Caffey syndrome. Nat Genet. 2002;32(3):448-52.
  104. Voloshin O, Gocheva Y, Gutnick M, Movshovich N, Bakhrat A, Baranes-Bachar K, et al. Tubulin chaperone E binds microtubules and proteasomes and protects against misfolded protein stress. Cellular and Molecular Life Sciences. 2010;67(12):2025-38.
  105. Parvari R, Diaz GA, and Hershkovitz E. Parathyroid Development and the Role of Tubulin Chaperone E. Hormone research in paediatrics. 2006;67(1):12-21.
  106. Chow J, Rahman J, Achermann JC, Dattani MT, and Rahman S. Mitochondrial disease and endocrine dysfunction. Nature Reviews Endocrinology. 2016;13(2):92-104.
  107. Seneca S, Meirleir LD, Scbepper JD, Balduck N, Jochmans K, Liebaers I, et al. Pearson marrow pancreas syndrome: a molecular study and clinical management. Clinical genetics. 1997;51(5):338-42.
  108. Tengan CH. Mitochondrial Encephalomyopathy and Hypoparathyrodism Associated with a Duplication and a Deletion of Mitochondrial Deoxyribonucleic Acid. Journal of Clinical Endocrinology &amp; Metabolism. 1998;83(1):125-9.
  109. Wilichowski E, Gruters A, Kruse K, Rating D, Beetz R, Korenke GC, et al. Hypoparathyroidism and Deafness Associated with Pleioplasmic Large Scale Rearrangements of the Mitochondrial DNA: A Clinical and Molecular Genetic Study of Four Children with Kearns-Sayre Syndrome. Pediatric Research. 1997;41(2):193-200.
  110. Naiki M, Ochi N, Kato YS, Purevsuren J, Yamada K, Kimura R, et al. Mutations in <i>HADHB</i>, which encodes the β‐subunit of mitochondrial trifunctional protein, cause infantile onset hypoparathyroidism and peripheral polyneuropathy. American Journal of Medical Genetics Part A. 2014;164(5):1180-7.
  111. Tyni T, Rapola J, Palotie A, and Pihko H. Hypoparathyroidism in a patient with long-chain 3-hydroxyacyl-coenzyme A dehydrogenase deficiency caused by the G1528C mutation. The Journal of Pediatrics. 1997;131(5):766-8.
  112. Carpenter TO, Carnes DL, and Anast CS. Hypoparathyroidism in Wilson's Disease. New England Journal of Medicine. 1983;309(15):873-7.
  113. Brown EM. Anti-parathyroid and anti-calcium sensing receptor antibodies in autoimmune hypoparathyroidism. Endocrinol Metab Clin North Am. 2009;38(2):437-45, x.
  114. Kemp EH, and Weetman AP. Hypoparathyroidism. Springer Milan; 2015:177-88.
  115. Alimohammadi M, Bjorklund P, Hallgren A, Pontynen N, Szinnai G, Shikama N, et al. Autoimmune Polyendocrine Syndrome Type 1 and NALP5, a Parathyroid Autoantigen. New England Journal of Medicine. 2008;358(10):1018-28.
  116. Eisenbarth GS, and Gottlieb PA. Autoimmune Polyendocrine Syndromes. New England Journal of Medicine. 2004;350(20):2068-79.
  117. Li Y, Song YH, Rais N, Connor E, Schatz D, Muir A, et al. Autoantibodies to the extracellular domain of the calcium sensing receptor in patients with acquired hypoparathyroidism. The Journal of clinical investigation. 1996;97(4):910-4.
  118. Goswami R, Brown EM, Kochupillai N, Gupta N, Rani R, Kifor O, et al. Prevalence of calcium sensing receptor autoantibodies in patients with sporadic idiopathic hypoparathyroidism. European Journal of Endocrinology. 2004:9-18.
  119. Gylling M, Kääriäinen E, Väisänen R, Kerosuo L, Solin M-L, Halme L, et al. The Hypoparathyroidism of Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy Protective Effect of Male Sex. The Journal of Clinical Endocrinology &amp; Metabolism. 2003;88(10):4602-8.
  120. Kemp EH, Gavalas NG, Krohn KJE, Brown EM, Watson PF, and Weetman AP. Activating autoantibodies against the calcium-sensing receptor detected in two patients with autoimmune polyendocrine syndrome type 1. The Journal of clinical endocrinology and metabolism. 2009;94(12):4749-56.
  121. Soderbergh A, Myhre AG, Ekwall O, Gebre-Medhin G, Hedstrand H, Landgren E, et al. Prevalence and Clinical Associations of 10 Defined Autoantibodies in Autoimmune Polyendocrine Syndrome Type I. The Journal of Clinical Endocrinology &amp; Metabolism. 2004;89(2):557-62.
  122. Mayer A, Ploix C, Orgiazzi J, Desbos A, Moreira A, Vidal H, et al. Calcium-Sensing Receptor Autoantibodies Are Relevant Markers of Acquired Hypoparathyroidism. The Journal of Clinical Endocrinology &amp; Metabolism. 2004;89(9):4484-8.
  123. Schott M, and Scherbaum WA. Hypoparathyroidism and autoimmune polyendocrine syndromes. N Engl J Med. 2004;351(10):1032-3; author reply -3.
  124. Kifor O, McElduff A, LeBoff MS, Moore FD, Butters R, Gao P, et al. Activating Antibodies to the Calcium-Sensing Receptor in Two Patients with Autoimmune Hypoparathyroidism. The Journal of Clinical Endocrinology &amp; Metabolism. 2004;89(2):548-56.
  125. Buzi F, Badolato R, Mazza C, Giliani S, Notarangelo LD, Radetti G, et al. Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy Syndrome: Time to Review Diagnostic Criteria? The Journal of Clinical Endocrinology &amp; Metabolism. 2003;88(7):3146-8.
  126. Ferre EM, Rose SR, Rosenzweig SD, Burbelo PD, Romito KR, Niemela JE, et al. Redefined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI Insight. 2016;1(13).
  127. Halonen M, Kangas H, Rüppell T, Ilmarinen T, Ollila J, Kolmer M, et al. APECED-causing mutations in AIRE reveal the functional domains of the protein. Human Mutation. 2004;23(3):245-57.
  128. Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, et al. Positional cloning of the APECED gene. Nature Genetics. 1997;17(4):393-8.
  129. Aaltonen J, Bjorses P, Perheentupa J, Horelli–Kuitunen N, Palotie A, Peltonen L, et al. An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nature Genetics. 1997;17(4):399-403.
  130. Akirav EM, Ruddle NH, and Herold KC. The role of AIRE in human autoimmune disease. Nature Reviews Endocrinology. 2010;7(1):25-33.
  131. Cervato S, Mariniello B, Lazzarotto F, Morlin L, Zanchetta R, Radetti G, et al. Evaluation of the autoimmune regulator (AIRE) gene mutations in a cohort of Italian patients with autoimmune-polyendocrinopathy-candidiasis-ectodermal-dystrophy (APECED) and in their relatives. Clinical endocrinology. 2009;70(3):421-8.
  132. Cheng MH, Fan U, Grewal N, Barnes M, Mehta A, Taylor S, et al. Acquired autoimmune polyglandular syndrome, thymoma, and an AIRE defect. The New England journal of medicine. 2010;362(8):764-6.
  133. Chase LR, Melson GL, and Aurbach GD. Pseudohypoparathyroidism: defective excretion of 3',5'-AMP in response to parathyroid hormone. J Clin Invest. 1969;48:1832-44.
  134. Albright F, Burnett CH, Smith PH, and Parson W. Pseudohypoparathyroidism - an example of "Seabright-Bantam syndrome". Endocrinology. 1942;30:922-32.
  135. Wilson LC, and Trembath RC. Albright's hereditary osteodystrophy. J Med Genet. 1994;31(10):779-84.
  136. de Sanctis L, Vai S, Andreo MR, Romagnolo D, Silvestro L, and de Sanctis C. Brachydactyly in 14 genetically characterized pseudohypoparathyroidism type Ia patients. J Clin Endocrinol Metab. 2004;89(4):1650-5.
  137. Puzhko S, Goodyer CG, Mohammad AK, Canaff L, Misra M, Jüppner H, et al. Parathyroid hormone signaling via Galphas is selectively inhibited by an NH(2) -terminally truncated Galphas: Implications for pseudohypoparathyroidism. J Bone Miner Res. 2011;26(10):2473-85.
  138. Poznanski AK, Werder EA, Giedion A, Martin A, and Shaw H. The pattern of shortening of the bones of the hand in PHP and PPHP--A comparison with brachydactyly E, Turner Syndrome, and acrodysostosis. Radiology. 1977;123(3):707-18.
  139. Levine MA. Clinical spectrum and pathogenesis of pseudohypoparathyroidism. Reviews in endocrine & metabolic disorders. 2000;1(4):265-74.
  140. Brickman AS, Stern N, and Sowers JR. Hypertension in pseudohypoparathyroidism type I. Am J Med. 1988;85(6):785-92.
  141. Koch T, Lehnhardt E, Bottinger H, Pfeuffer T, Palm D, Fischer B, et al. Sensorineural hearing loss owing to deficient G proteins in patients with pseudohypoparathyroidism: results of a multicentre study. Eur J Clin Invest. 1990;20(4):416-21.
  142. Goadsby PJ, Lollin Y, and Kocen RS. Pseudopseudohypoparathyroidism and spinal cord compression. J Neurol Neurosurg Psychiatry. 1991;54(10):929-31.
  143. Maeda SS, Fortes EM, Oliveira UM, Borba VC, and Lazaretti-Castro M. Hypoparathyroidism and pseudohypoparathyroidism. Arq Bras Endocrinol Metabol. 2006;50(4):664-73.
  144. Ong KK, Amin R, and Dunger DB. Pseudohypoparathyroidism--another monogenic obesity syndrome. Clinical endocrinology. 2000;52(3):389-91.
  145. Chen M, Shrestha YB, Podyma B, Cui Z, Naglieri B, Sun H, et al. Gsalpha deficiency in the dorsomedial hypothalamus underlies obesity associated with Gsalpha mutations. J Clin Invest. 2017;127(2):500-10.
  146. Chen M, Wang J, Dickerson KE, Kelleher J, Xie T, Gupta D, et al. Central nervous system imprinting of the G protein G(s)alpha and its role in metabolic regulation. Cell Metab. 2009;9(6):548-55.
  147. Mendes de Oliveira E, Keogh JM, Talbot F, Henning E, Ahmed R, Perdikari A, et al. Obesity-Associated GNAS Mutations and the Melanocortin Pathway. N Engl J Med. 2021;385(17):1581-92.
  148. Phelan MC, Rogers RC, Clarkson KB, Bowyer FP, Levine MA, Estabrooks LL, et al. Albright hereditary osteodystrophy and del(2) (q37.3) in four unrelated individuals. American journal of medical genetics. 1995;58(1):1-7.
  149. Williams SR, Aldred MA, Der Kaloustian VM, Halal F, Gowans G, McLeod DR, et al. Haploinsufficiency of HDAC4 causes brachydactyly mental retardation syndrome, with brachydactyly type E, developmental delays, and behavioral problems. Am J Hum Genet. 2010;87(2):219-28.
  150. Johnson D, Kan SH, Oldridge M, Trembath RC, Roche P, Esnouf RM, et al. Missense mutations in the homeodomain of HOXD13 are associated with brachydactyly types D and E. Am J Hum Genet. 2003;72(4):984-97.
  151. Maass PG, Wirth J, Aydin A, Rump A, Stricker S, Tinschert S, et al. A cis-regulatory site downregulates PTHLH in translocation t(8;12)(q13;p11.2) and leads to Brachydactyly Type E. Hum Mol Genet. 2010;19(5):848-60.
  152. Klopocki E, Hennig BP, Dathe K, Koll R, de Ravel T, Baten E, et al. Deletion and point mutations of PTHLH cause brachydactyly type E. Am J Hum Genet. 2010;86(3):434-9.
  153. Maass PG, Rump A, Schulz H, Stricker S, Schulze L, Platzer K, et al. A misplaced lncRNA causes brachydactyly in humans. J Clin Invest. 2012;122(11):3990-4002.
  154. Maass PG, Aydin A, Luft FC, Schachterle C, Weise A, Stricker S, et al. PDE3A mutations cause autosomal dominant hypertension with brachydactyly. Nat Genet. 2015;47(6):647-53.
  155. Izzi B, Francois I, Labarque V, Thys C, Wittevrongel C, Devriendt K, et al. Methylation defect in imprinted genes detected in patients with an Albright's hereditary osteodystrophy like phenotype and platelet Gs hypofunction. PLoS One. 2012;7(6):e38579.
  156. Farfel Z, Brickman AS, Kaslow HR, Brothers VM, and Bourne HR. Defect of receptor-cyclase coupling protein in pseudohypoparathyroidism. N Engl J Med. 1980;303:237-42.
  157. Levine MA, Downs RW, Jr., Singer M, Marx SJ, Aurbach GD, and Spiegel AM. Deficient activity of guanine nucleotide regulatory protein in erythrocytes from patients with pseudohypoparathyroidism. Biochem Biophys Res Commun. 1980;94:1319-24.
  158. Levine MA, Jap TS, Mauseth RS, R.S. Downs J, and Spiegel AM. Activity of the stimulatory guanine nucleotide-binding protein is reduced in erythrocytes from patients with pseudohypoparathyroidism and pseudohypoparathyroidism:Biochemical, endocrine, and genetic analysis of Albright's hereditary osteodystrophy in six kindreds. J Clin Endocrinol Metab. 1986;62:497-502.
  159. Beaudet AL. Complex imprinting. Nat Genet. 2004;36(8):793-5.
  160. Yu S, Yu D, Lee E, Eckhaus M, Lee R, Corria Z, et al. Variable and tissue-specific hormone resistance in heterotrimeric Gs protein a-subunit (Gsa) knockout mice is due to tissue-specific imprinting of the Gsa gene. Proc Natl Acad Sci USA. 1998;95:8715-20.
  161. Hayward BE, Barlier A, Korbonits M, Grossman AB, Jacquet P, Enjalbert A, et al. Imprinting of the G(s)alpha gene GNAS1 in the pathogenesis of acromegaly. J Clin Invest. 2001;107(6):R31-6.
  162. Mantovani G, Ballare E, Giammona E, Beck-Peccoz P, and Spada A. The Gsalpha Gene: Predominant Maternal Origin of Transcription in Human Thyroid Gland and Gonads. J Clin Endocrinol Metab. 2002;87(10):4736-40.
  163. Germain-Lee EL, Ding CL, Deng Z, Crane JL, Saji M, Ringel MD, et al. Paternal imprinting of Galpha(s) in the human thyroid as the basis of TSH resistance in pseudohypoparathyroidism type 1a. Biochem Biophys Res Commun. 2002;296(1):67-72.
  164. Liu J, Erlichman B, and Weinstein LS. The stimulatory G protein a-subunit Gsa is imprinted in human thyroid glands: implications for thyroid function in pseudohypoparathyroidism types 1A and 1B. J Clin Endocrinol Metabol. 2003;88(9):4336-41.
  165. Davies AJ, and Hughes HE. Imprinting in Albright's hereditary osteodystrophy. J Med Genet. 1993;30:101-3.
  166. Liu J, Litman D, Rosenberg M, Yu S, Biesecker L, and Weinstein L. A GNAS1 imprinting defect in pseudohypoparathyroidism type IB. J Clin Invest. 2000;106:1167-74.
  167. Thiele S, Mantovani G, Barlier A, Boldrin V, Bordogna P, De Sanctis L, et al. From pseudohypoparathyroidism to inactivating PTH/PTHrP signalling disorder (iPPSD), a novel classification proposed by the EuroPHP network. Eur J Endocrinol. 2016;175(6):P1-P17.
  168. Zheng H, Radeva G, McCann JA, Hendy GN, and Goodyer CG. Gas transcripts are biallelically expressed in the human kidney cortex: implications for pseudohypoparathyroidism type Ib. J Clin Endocrinol Metab. 2001;86(10):4627-9.
  169. Weinstein LS, Liu J, Sakamoto A, Xie T, and Chen M. Minireview: GNAS: normal and abnormal functions. Endocrinology. 2004;145(12):5459-64.
  170. Weinstein LS, Yu S, and Ecelbarger CA. Variable imprinting of the heterotrimeric G protein G(s) alpha-subunit within different segments of the nephron. Am J Physiol Renal Physiol. 2000;278(4):F507-14.
  171. Turan S, Fernandez-Rebollo E, Aydin C, Zoto T, Reyes M, Bounoutas G, et al. Postnatal establishment of allelic Galphas silencing as a plausible explanation for delayed onset of parathyroid hormone resistance owing to heterozygous Galphas disruption. J Bone Miner Res. 2014;29(3):749-60.
  172. Weinstein LS, Gejman PV, Friedman E, Kadowaki T, Collins RM, Gershon ES, et al. Mutations of the Gs alpha-subunit gene in Albright hereditary osteodystrophy detected by denaturing gradient gel electrophoresis. Proc Natl Acad Sci U S A. 1990;87(21):8287-90.
  173. Patten JL, Johns DR, Valle D, Eil C, Gruppuso PA, Steele G, et al. Mutation in the gene encoding the stimulatory G protein of adenylate cyclase in Albright's hereditary osteodystrophy. New Engl J Med. 1990;322:1412-9.
  174. Weinstein LS, Gejman PV, de Mazancourt P, American N, and Spiegel AM. A heterozygous 4-bp deletion mutation in the Gs alpha gene (GNAS1) in a patient with Albright hereditary osteodystrophy. Genomics. 1992;13(4):1319-21.
  175. Ahrens W, Hiort O, Staedt P, Kirschner T, Marschke C, and Kruse K. Analysis of the GNAS1 gene in Albright's hereditary osteodystrophy. J Clin Endocrinol Metab. 2001;86(10):4630-4.
  176. Aldred MA, Bagshaw RJ, Macdermot K, Casson D, Murch SH, Walker-Smith JA, et al. Germline mosaicism for a GNAS1 mutation and Albright hereditary osteodystrophy. J Med Genet. 2000;37(11):E35.
  177. Fernandez-Rebollo E, Garcia-Cuartero B, Garin I, Largo C, Martinez F, Garcia-Lacalle C, et al. Intragenic GNAS deletion involving exon A/B in pseudohypoparathyroidism type 1A resulting in an apparent loss of exon A/B methylation: potential for misdiagnosis of pseudohypoparathyroidism type 1B. J Clin Endocrinol Metab. 2010;95(2):765-71.
  178. Levine MA. An update on the clinical and molecular characteristics of pseudohypoparathyroidism. Current opinion in endocrinology, diabetes, and obesity. 2012;19(6):443-51.
  179. Namnoum AB, Merriam GR, Moses AM, and Levine MA. Reproductive dysfunction in women with Albright's hereditary osteodystrophy. J Clin Endocrinol Metab. 1998;83:824-9.
  180. McIlroy J, Dryburgh F, Hinnie J, Dargie R, and Al-Rawi A. Oestrogen and calcium homeostasis in women with hypoparathyroidism. BMJ. 1999;319(7219):1252-3.
  181. Breslau NA, and Zerwekh JE. Relationship of estrogen and pregnancy to calcium homeostasis in pseudohypoparathyroidism. J Clin Endocrinol Metab. 1986;62(1):45-51.
  182. Germain-Lee EL, Groman J, Crane JL, Jan de Beur SM, and Levine MA. Growth hormone deficiency in pseudohypoparathyroidism type 1a: another manifestation of multihormone resistance. J Clin Endocrinol Metab. 2003;88(9):4059-69.
  183. Mantovani G, Maghnie M, Weber G, De Menis E, Brunelli V, Cappa M, et al. Growth hormone-releasing hormone resistance in pseudohypoparathyroidism type ia: new evidence for imprinting of the Gs alpha gene. J Clin Endocrinol Metab. 2003;88(9):4070-4.
  184. Long DN, McGuire S, Levine MA, Weinstein LS, and Germain-Lee EL. Body mass index differences in pseudohypoparathyroidism type 1a versus pseudopseudohypoparathyroidism may implicate paternal imprinting of Galpha(s) in the development of human obesity. J Clin Endocrinol Metab. 2007;92(3):1073-9.
  185. Mantovani G, Bondioni S, Locatelli M, Pedroni C, Lania AG, Ferrante E, et al. Biallelic expression of the Gsalpha gene in human bone and adipose tissue. J Clin Endocrinol Metab. 2004;89(12):6316-9.
  186. Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, and O'Rahilly S. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet. 1998;20(2):111-2.
  187. Vaisse C, Clement K, Guy-Grand B, and Froguel P. A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nat Genet. 1998;20(2):113-4.
  188. Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88(1):131-41.
  189. Mouallem M, Shaharabany M, Weintrob N, Shalitin S, Nagelberg N, Shapira H, et al. Cognitive impairment is prevalent in pseudohypoparathyroidism type Ia, but not in pseudopseudohypoparathyroidism: possible cerebral imprinting of Gsalpha. Clinical endocrinology. 2008;68(2):233-9.
  190. Peterman MG, and Garvey JL. Pseudohypoparathyroidsim; case report. Pediatrics. 1949;4(6):790-7, illust.
  191. Reynolds TB, Jacobson G, Edmondson HA, Martin HE, and Nelson CH. Pseudohypoparathyroidism; report of a case showing bony demineralization. J Clin Endocrinol Metab. 1952;12(5):560-73.
  192. Farfel Z, Brothers VM, Brickman AS, Conte F, Neer R, and Bourne HR. Pseudohypoparathyroidism: inheritance of deficient receptor-cyclase coupling activity. Proc Natl Acad Sci U S A. 1981;78(5):3098-102.
  193. Ish-Shalom S, Rao LG, Levine MA, Fraser D, Kooh SW, Josse RG, et al. Normal parathyroid hormone responsiveness of bone-derived cells from a patient with pseudohypoparathyroidism. J Bone Miner Res. 1996;11:8-14.
  194. Murray T, Gomez Rao E, Wong MM, Waddell JP, McBroom R, Tam CS, et al. Pseudohypoparathyroidism with osteitis fibrosa cystica:direct demonstration of skeletal responsiveness to parathyroid hormone in cells cultured from bone. J Bone Miner Res. 1993;8:83-91.
  195. Farfel Z. Pseudohypohyperparathyroidism-pseudohypoparathyroidism type Ib. J Bone Miner Res. 1999;14:1016.
  196. Molinaro A, Tiosano D, Takatani R, Chrysis D, Russell W, Koscielniak N, et al. TSH elevations as the first laboratory evidence for pseudohypoparathyroidism type Ib (PHP-Ib). J Bone Miner Res. 2015;30(5):906-12.
  197. Romanet P, Osei L, Netchine I, Pertuit M, Enjalbert A, Reynaud R, et al. Case report of GNAS epigenetic defect revealed by a congenital hypothyroidism. Pediatrics. 2015;135(4):e1079-83.
  198. Sano S, Iwata H, Matsubara K, Fukami M, Kagami M, and Ogata T. Growth hormone deficiency in monozygotic twins with autosomal dominant pseudohypoparathyroidism type Ib. Endocrine journal. 2015;62(6):523-9.
  199. Laspa E, Bastepe M, Jüppner H, and Tsatsoulis A. Phenotypic and molecular genetic aspects of pseudohypoparathyroidism type ib in a Greek kindred: evidence for enhanced uric acid excretion due to parathyroid hormone resistance. J Clin Endocrinol Metab. 2004;89(12):5942-7.
  200. Unluturk U, Harmanci A, Babaoglu M, Yasar U, Varli K, Bastepe M, et al. Molecular diagnosis and clinical characterization of pseudohypoparathyroidism type-Ib in a patient with mild Albright's hereditary osteodystrophy-like features, epileptic seizures, and defective renal handling of uric acid. Am J Med Sci. 2008;336(1):84-90.
  201. Schipani E, Weinstein LS, Bergwitz C, Iida-Klein A, Kong XF, Stuhrmann M, et al. Pseudohypoparathyroidism type Ib is not caused by mutations in the coding exons of the human parathyroid hormone (PTH)/PTH-related peptide receptor gene. J Clin Endocrinol Metab. 1995;80:1611-21.
  202. Fukumoto S, Suzawa M, Takeuchi Y, Nakayama K, Kodama Y, Ogata E, et al. Absence of mutations in parathyroid hormone (PTH)/PTH-related protein receptor complementary deoxyribonucleic acid in patients with pseudohypoparathyroidism type Ib. J Clin Endocrinol Metab. 1996;81:2554-8.
  203. Bettoun JD, Minagawa M, Kwan MY, Lee HS, Yasuda T, Hendy GN, et al. Cloning and characterization of the promoter regions of the human parathyroid hormone (PTH)/PTH-related peptide receptor gene:analysis of deoxyribonucleic acid from normal subjects and patients with pseudohypoparathyroidism type Ib. J Clin Endocrinol Metab. 1997;82:1031-40.
  204. Jüppner H, Schipani E, Bastepe M, Cole DEC, Lawson ML, Mannstadt M, et al. The gene responsible for pseudohypoparathyroidism type Ib is paternally imprinted and maps in four unrelated kindreds to chromosome 20q13.3. Proc Natl Acad Sci USA. 1998;95:11798-803.
  205. Jan de Beur S, Ding C, LaBuda M, Usdin T, and Levine M. Pseudohypoparathyroidism 1b: exclusion of parathyroid hormone and its receptors as candidate disease genes. J Clin Endocrinol Metab. 2000;85:2239-46.
  206. Bastepe M, Pincus JE, Sugimoto T, Tojo K, Kanatani M, Azuma Y, et al. Positional dissociation between the genetic mutation responsible for pseudohypoparathyroidism type Ib and the associated methylation defect at exon A/B: evidence for a long-range regulatory element within the imprinted GNAS1 locus. Hum Mol Genet. 2001;10:1231-41.
  207. Bastepe M, Fröhlich LF, Hendy GN, Indridason OS, Josse RG, Koshiyama H, et al. Autosomal dominant pseudohypoparathyroidism type Ib is associated with a heterozygous microdeletion that likely disrupts a putative imprinting control element of GNAS. J Clin Invest. 2003;112(8):1255-63.
  208. Linglart A, Gensure RC, Olney RC, Jüppner H, and Bastepe M. A Novel STX16 Deletion in Autosomal Dominant Pseudohypoparathyroidism Type Ib Redefines the Boundaries of a cis-Acting Imprinting Control Element of GNAS. Am J Hum Genet. 2005;76(5):804-14.
  209. Elli FM, de Sanctis L, Peverelli E, Bordogna P, Pivetta B, Miolo G, et al. Autosomal dominant pseudohypoparathyroidism type Ib: a novel inherited deletion ablating STX16 causes loss of imprinting at the A/B DMR. J Clin Endocrinol Metab. 2014;99(4):E724-8.
  210. Yang Y, Chu X, Nie M, Song A, Jiang Y, Li M, et al. A novel long-range deletion spanning STX16 and NPEPL1 causing imprinting defects of the GNAS locus discovered in a patient with autosomal-dominant pseudohypoparathyroidism type 1B. Endocrine. 2020;69(1):212-9.
  211. Danzig J, Li D, Jan de Beur S, and Levine MA. High-throughput Molecular Analysis of Pseudohypoparathyroidism 1b Patients Reveals Novel Genetic and Epigenetic Defects. J Clin Endocrinol Metab. 2021;106(11):e4603-e20.
  212. Richard N, Abeguile G, Coudray N, Mittre H, Gruchy N, Andrieux J, et al. A new deletion ablating NESP55 causes loss of maternal imprint of A/B GNAS and autosomal dominant pseudohypoparathyroidism type Ib. J Clin Endocrinol Metab. 2012;97(5):E863-7.
  213. Bastepe M, Fröhlich LF, Linglart A, Abu-zahra HS, Tojo K, Ward LM, et al. Deletion of the NESP55 differentially methylated region causes loss of maternal GNAS imprints and pseudohypoparathyroidism type-Ib. Nat Genet. 2005;37(1):25-37.
  214. Chillambhi S, Turan S, Hwang DY, Chen HC, Jüppner H, and Bastepe M. Deletion of the Noncoding GNAS Antisense Transcript Causes Pseudohypoparathyroidism Type Ib and Biparental Defects of GNAS Methylation in cis. J Clin Endocrinol Metab. 2010;95(8):3993-4002.
  215. Rezwan FI, Poole RL, Prescott T, Walker JM, Karen Temple I, and Mackay DJ. Very small deletions within the NESP55 gene in pseudohypoparathyroidism type 1b. Eur J Hum Genet. 2015;23(4):494-9.
  216. Perez-Nanclares G, Velayos T, Vela A, Munoz-Torres M, and Castano L. Pseudohypoparathyroidism type Ib associated with novel duplications in the GNAS locus. PLoS One. 2015;10(2):e0117691.
  217. Nakamura A, Hamaguchi E, Horikawa R, Nishimura Y, Matsubara K, Sano S, et al. Complex Genomic Rearrangement Within the GNAS Region Associated With Familial Pseudohypoparathyroidism Type 1b. J Clin Endocrinol Metab. 2016;101(7):2623-7.
  218. Grigelioniene G, Nevalainen PI, Reyes M, Thiele S, Tafaj O, Molinaro A, et al. A Large Inversion Involving GNAS Exon A/B and All Exons Encoding Gsalpha Is Associated With Autosomal Dominant Pseudohypoparathyroidism Type Ib (PHP1B). J Bone Miner Res. 2017;32(4):776-83.
  219. Takatani R, Molinaro A, Grigelioniene G, Tafaj O, Watanabe T, Reyes M, et al. Analysis of Multiple Families With Single Individuals Affected by Pseudohypoparathyroidism Type Ib (PHP1B) Reveals Only One Novel Maternally Inherited GNAS Deletion. J Bone Miner Res. 2016;31(4):796-805.
  220. Fröhlich LF, Bastepe M, Ozturk D, Abu-Zahra H, and Jüppner H. Lack of Gnas epigenetic changes and pseudohypoparathyroidism type Ib in mice with targeted disruption of syntaxin-16. Endocrinology. 2007;148(6):2925-35.
  221. Yang H, Bai D, Li Y, Yu Z, Wang C, Sheng Y, et al. Allele-specific H3K9me3 and DNA methylation co-marked CpG-rich regions serve as potential imprinting control regions in pre-implantation embryo. Nature cell biology. 2022;24(5):783-92.
  222. Yagi M, Kabata M, Ukai T, Ohta S, Tanaka A, Shimada Y, et al. De Novo DNA Methylation at Imprinted Loci during Reprogramming into Naive and Primed Pluripotency. Stem cell reports. 2019;12(5):1113-28.
  223. Iwasaki Y, Aksu C, Reyes M, Ay B, He Q, and Bastepe M. The long-range interaction between two GNAS imprinting control regions delineates pseudohypoparathyroidism type 1B pathogenesis. J Clin Invest. 2023;133(8):e167953.
  224. Chotalia M, Smallwood SA, Ruf N, Dawson C, Lucifero D, Frontera M, et al. Transcription is required for establishment of germline methylation marks at imprinted genes. Genes Dev. 2009;23(1):105-17.
  225. Bastepe M, Lane AH, and Jüppner H. Paternal uniparental isodisomy of chromosome 20q (patUPD20q) - and the resulting changes in GNAS1 methylation - as a plausible cause of pseudohypoparathyroidism. Am J Hum Genet. 2001;68:1283-9.
  226. Lecumberri B, Fernandez-Rebollo E, Sentchordi L, Saavedra P, Bernal-Chico A, Pallardo LF, et al. Coexistence of two different pseudohypoparathyroidism subtypes (Ia and Ib) in the same kindred with independent Gs{alpha} coding mutations and GNAS imprinting defects. J Med Genet. 2010;47(4):276-80.
  227. Fernandez-Rebollo E, Lecumberri B, Garin I, Arroyo J, Bernal-Chico A, Goni F, et al. New mechanisms involved in paternal 20q disomy associated with pseudohypoparathyroidism. Eur J Endocrinol. 2010;163(6):953-62.
  228. Dixit A, Chandler KE, Lever M, Poole RL, Bullman H, Mughal MZ, et al. Pseudohypoparathyroidism type 1b due to paternal uniparental disomy of chromosome 20q. J Clin Endocrinol Metab. 2013;98(1):E103-8.
  229. Takatani R, Minagawa M, Molinaro A, Reyes M, Kinoshita K, Takatani T, et al. Similar frequency of paternal uniparental disomy involving chromosome 20q (patUPD20q) in Japanese and Caucasian patients affected by sporadic pseudohypoparathyroidism type Ib (sporPHP1B). Bone. 2015;79:15-20.
  230. Maupetit-Mehouas S, Azzi S, Steunou V, Sakakini N, Silve C, Reynes C, et al. Simultaneous hyper- and hypomethylation at imprinted loci in a subset of patients with GNAS epimutations underlies a complex and different mechanism of multilocus methylation defect in pseudohypoparathyroidism type 1b. Hum Mutat. 2013;34(8):1172-80.
  231. Bliek J, Verde G, Callaway J, Maas SM, De Crescenzo A, Sparago A, et al. Hypomethylation at multiple maternally methylated imprinted regions including PLAGL1 and GNAS loci in Beckwith-Wiedemann syndrome. Eur J Hum Genet. 2009;17(5):611-9.
  232. Mackay DJ, Callaway JL, Marks SM, White HE, Acerini CL, Boonen SE, et al. Hypomethylation of multiple imprinted loci in individuals with transient neonatal diabetes is associated with mutations in ZFP57. Nat Genet. 2008;40(8):949-51.
  233. Bakker B, Sonneveld LJ, Woltering MC, Bikker H, and Kant SG. A Girl With Beckwith-Wiedemann Syndrome and Pseudohypoparathyroidism Type 1B Due to Multiple Imprinting Defects. J Clin Endocrinol Metab. 2015;100(11):3963-6.
  234. Rochtus A, Martin-Trujillo A, Izzi B, Elli F, Garin I, Linglart A, et al. Genome-wide DNA methylation analysis of pseudohypoparathyroidism patients with GNAS imprinting defects. Clin Epigenetics. 2016;8:10.
  235. Hanna P, Francou B, Delemer B, Juppner H, and Linglart A. A Novel Familial PHP1B Variant With Incomplete Loss of Methylation at GNAS-A/B and Enhanced Methylation at GNAS-AS2. J Clin Endocrinol Metab. 2021;106(9):2779-87.
  236. Kawashima S, Yuno A, Sano S, Nakamura A, Ishiwata K, Kawasaki T, et al. Familial Pseudohypoparathyroidism Type IB Associated with an SVA Retrotransposon Insertion in the GNAS Locus. J Bone Miner Res. 2022;37(10):1850-9.
  237. Miller DE, Hanna P, Galey M, Reyes M, Linglart A, Eichler EE, et al. Targeted Long-Read Sequencing Identifies a Retrotransposon Insertion as a Cause of Altered GNAS Exon A/B Methylation in a Family With Autosomal Dominant Pseudohypoparathyroidism Type 1b (PHP1B). J Bone Miner Res. 2022;37(9):1711-9.
  238. Linglart A, Bastepe M, and Jüppner H. Similar clinical and laboratory findings in patients with symptomatic autosomal dominant and sporadic pseudohypoparathyroidism type Ib despite different epigenetic changes at the GNAS locus. Clinical endocrinology. 2007;67(6):822-31.
  239. Brehin AC, Colson C, Maupetit-Mehouas S, Grybek V, Richard N, Linglart A, et al. Loss of methylation at GNAS exon A/B is associated with increased intrauterine growth. J Clin Endocrinol Metab. 2015;100(4):E623-31.
  240. de Nanclares GP, Fernandez-Rebollo E, Santin I, Garcia-Cuartero B, Gaztambide S, Menendez E, et al. Epigenetic defects of GNAS in patients with pseudohypoparathyroidism and mild features of Albright's hereditary osteodystrophy. J Clin Endocrinol Metab. 2007;92(6):2370-3.
  241. Mariot V, Maupetit-Mehouas S, Sinding C, Kottler ML, and Linglart A. A maternal epimutation of GNAS leads to Albright osteodystrophy and parathyroid hormone resistance. J Clin Endocrinol Metab. 2008;93(3):661-5.
  242. Mantovani G, de Sanctis L, Barbieri AM, Elli FM, Bollati V, Vaira V, et al. Pseudohypoparathyroidism and GNAS epigenetic defects: clinical evaluation of albright hereditary osteodystrophy and molecular analysis in 40 patients. J Clin Endocrinol Metab. 2010;95(2):651-8.
  243. Wu WI, Schwindinger WF, Aparicio LF, and Levine MA. Selective resistance to parathyroid hormone caused by a novel uncoupling mutation in the carboxyl terminus of Gas: A cause of pseudohypoparathyroidism type Ib. J Biol Chem. 2001;276(1):165-71.
  244. Linglart A, Carel JC, Garabedian M, Le T, Mallet E, and Kottler ML. GNAS1 Lesions in Pseudohypoparathyroidism Ia and Ic: Genotype Phenotype Relationship and Evidence of the Maternal Transmission of the Hormonal Resistance. J Clin Endocrinol Metab. 2002;87(1):189-97.
  245. Linglart A, Mahon MJ, Kerachian MA, Berlach DM, Hendy GN, Jüppner H, et al. Coding GNAS mutations leading to hormone resistance impair in vitro agonist- and cholera toxin-induced adenosine cyclic 3',5'-monophosphate formation mediated by human XLas. Endocrinology. 2006;147(5):2253-62.
  246. Thiele S, de Sanctis L, Werner R, Grotzinger J, Aydin C, Jüppner H, et al. Functional characterization of GNAS mutations found in patients with pseudohypoparathyroidism type Ic defines a new subgroup of pseudohypoparathyroidism affecting selectively Gsalpha-receptor interaction. Hum Mutat. 2011;32(6):653-60.
  247. Drezner M, Neelon FA, and Lebovitz HE. Pseudohypoparathyroidism type II: a possible defect in the reception of the cyclic AMP signal. N Engl J Med. 1973;289(20):1056-60.
  248. Van Dop C. Pseudohypoparathyroidism: clinical and molecular aspects. Seminars in nephrology. 1989;9(2):168-78.
  249. Kruse K, and Kustermann W. Evidence for transient peripheral resistance to parathyroid hormone in premature infants. Acta Paediatr Scand. 1987;76(1):115-8.
  250. Lee CT, Tsai WY, Tung YC, and Tsau YK. Transient pseudohypoparathyroidism as a cause of late-onset hypocalcemia in neonates and infants. J Formos Med Assoc. 2008;107(10):806-10.
  251. Manzar S. Transient pseudohypoparathyroidism and neonatal seizure. J Trop Pediatr. 2001;47(2):113-4.
  252. Linglart A, Menguy C, Couvineau A, Auzan C, Gunes Y, Cancel M, et al. Recurrent PRKAR1A mutation in acrodysostosis with hormone resistance. N Engl J Med. 2011;364(23):2218-26.
  253. Robinow M, Pfeiffer RA, Gorlin RJ, McKusick VA, Renuart AW, Johnson GF, et al. Acrodysostosis. A syndrome of peripheral dysostosis, nasal hypoplasia, and mental retardation. Am J Dis Child. 1971;121(3):195-203.
  254. Maroteaux P, and Malamut G. [Acrodysostosis]. La Presse medicale. 1968;76(46):2189-92.
  255. Linglart A, Fryssira H, Hiort O, Holterhus PM, Perez de Nanclares G, Argente J, et al. PRKAR1A and PDE4D mutations cause acrodysostosis but two distinct syndromes with or without GPCR-signaling hormone resistance. J Clin Endocrinol Metab. 2012;97(12):E2328-38.
  256. Nagasaki K, Iida T, Sato H, Ogawa Y, Kikuchi T, Saitoh A, et al. PRKAR1A mutation affecting cAMP-mediated G protein-coupled receptor signaling in a patient with acrodysostosis and hormone resistance. J Clin Endocrinol Metab. 2012;97(9):E1808-13.
  257. Silve C. Acrodysostosis: A new form of pseudohypoparathyroidism? Ann Endocrinol (Paris). 2015;76(2):110-2.
  258. Michot C, Le Goff C, Goldenberg A, Abhyankar A, Klein C, Kinning E, et al. Exome sequencing identifies PDE4D mutations as another cause of acrodysostosis. Am J Hum Genet. 2012;90(4):740-5.
  259. Bruystens JG, Wu J, Fortezzo A, Del Rio J, Nielsen C, Blumenthal DK, et al. Structure of a PKA RIalpha Recurrent Acrodysostosis Mutant Explains Defective cAMP-Dependent Activation. Journal of molecular biology. 2016;428(24 Pt B):4890-904.
  260. Le Stunff C, Tilotta F, Sadoine J, Le Denmat D, Briet C, Motte E, et al. Knock-In of the Recurrent R368X Mutation of PRKAR1A that Represses cAMP-Dependent Protein Kinase A Activation: A Model of Type 1 Acrodysostosis. J Bone Miner Res. 2017;32(2):333-46.
  261. Rhayem Y, Le Stunff C, Abdel Khalek W, Auzan C, Bertherat J, Linglart A, et al. Functional Characterization of PRKAR1A Mutations Reveals a Unique Molecular Mechanism Causing Acrodysostosis but Multiple Mechanisms Causing Carney Complex. J Biol Chem. 2015;290(46):27816-28.
  262. Lee H, Graham JM, Jr., Rimoin DL, Lachman RS, Krejci P, Tompson SW, et al. Exome sequencing identifies PDE4D mutations in acrodysostosis. Am J Hum Genet. 2012;90(4):746-51.
  263. Lynch DC, Dyment DA, Huang L, Nikkel SM, Lacombe D, Campeau PM, et al. Identification of novel mutations confirms PDE4D as a major gene causing acrodysostosis. Hum Mutat. 2013;34(1):97-102.
  264. Conti M, and Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem. 2007;76:481-511.
  265. Boda H, Uchida H, Takaiso N, Ouchi Y, Fujita N, Kuno A, et al. A PDE3A mutation in familial hypertension and brachydactyly syndrome. J Hum Genet. 2016;61(8):701-3.
  266. Weinstein LS. The Genetics of Osteoporosis and Metabolic Bone Disease. Humana Press; 2000:163-77.
  267. Weinstein LS, and Collins MT. Principles of Bone Biology. Elsevier; 2008:1453-77.
  268. Cole DE, Fraser FC, Glorieux FH, Jequier S, Marie PJ, Reade TM, et al. Panostotic fibrous dysplasia: a congenital disorder of bone with unusual facial appearance, bone fragility, hyperphosphatasemia, and hypophosphatemia. American journal of medical genetics. 1983;14(4):725-35.
  269. Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, and Vallar L. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature. 1989;340(6236):692-6.
  270. Iiri T, Herzmark P, Nakamoto JM, Dop Cv, and Bourne HR. Rapid GDP release from Gs in patients with gain and loss of function. Nature. 1994;371(6493):164-8.
  271. Nakamoto JM, Zimmerman D, Jones EA, Loke KY, Siddiq K, Donlan MA, et al. Concurrent hormone resistance (pseudohypoparathyroidism type Ia) and hormone independence (testotoxicosis) caused by a unique mutation in the G alpha s gene. Biochemical and molecular medicine. 1996;58(1):18-24.
  272. Makita N, Sato J, Rondard P, Fukamachi H, Yuasa Y, Aldred MA, et al. Human G(salpha) mutant causes pseudohypoparathyroidism type Ia/neonatal diarrhea, a potential cell-specific role of the palmitoylation cycle. Proc Natl Acad Sci U S A. 2007;104(44):17424-9.
  273. Wentworth K, Hsing A, Urrutia A, Zhu Y, Horvai AE, Bastepe M, et al. A Novel T55A Variant of Gs alpha Associated with Impaired cAMP Production, Bone Fragility, and Osteolysis. Case Rep Endocrinol. 2016;2016:2691385.
  274. Biebermann H, Kleinau G, Schnabel D, Bockenhauer D, Wilson LC, Tully I, et al. A New Multisystem Disorder Caused by the Galphas Mutation p.F376V. J Clin Endocrinol Metab. 2019;104(4):1079-89.
  275. Eddy MC, De Beur SM, Yandow SM, McAlister WH, Shore EM, Kaplan FS, et al. Deficiency of the alpha-subunit of the stimulatory G protein and severe extraskeletal ossification. J Bone Miner Res. 2000;15(11):2074-83.
  276. Kaplan FS, and Shore EM. Progressive osseous heteroplasia. J Bone Miner Res. 2000;15(11):2084-94.
  277. Shore EM, and Kaplan FS. Inherited human diseases of heterotopic bone formation. Nat Rev Rheumatol. 2010;6(9):518-27.
  278. Yeh GL, Mathur S, Wivel A, Li M, Gannon FH, Ulied A, et al. GNAS1 mutation and Cbfa1 misexpression in a child with severe congenital platelike osteoma cutis. J Bone Miner Res. 2000;15(11):2063-73.
  279. Adegbite NS, Xu M, Kaplan FS, Shore EM, and Pignolo RJ. Diagnostic and mutational spectrum of progressive osseous heteroplasia (POH) and other forms of GNAS-based heterotopic ossification. Am J Med Genet A. 2008;146A(14):1788-96.
  280. Shore EM, Ahn J, Jan de Beur S, Li M, Xu M, Gardner RJ, et al. Paternally inherited inactivating mutations of the GNAS1 gene in progressive osseous heteroplasia. N Engl J Med. 2002;346(2):99-106.
  281. Ahmed SF, Barr DG, and Bonthron DT. GNAS1 mutations and progressive osseous heteroplasia. N Engl J Med. 2002;346(21):1669-71.
  282. Cairns DM, Pignolo RJ, Uchimura T, Brennan TA, Lindborg CM, Xu M, et al. Somitic disruption of GNAS in chick embryos mimics progressive osseous heteroplasia. J Clin Invest. 2013;123(8):3624-33.
  283. Gardella TJ, and Vilardaga JP. International Union of Basic and Clinical Pharmacology. XCIII. The parathyroid hormone receptors--family B G protein-coupled receptors. Pharmacol Rev. 2015;67(2):310-37.
  284. Scillitani A, Jang C, Wong BY, Hendy GN, and Cole DE. A functional polymorphism in the PTHR1 promoter region is associated with adult height and BMD measured at the femoral neck in a large cohort of young caucasian women. Hum Genet. 2006;119(4):416-21.
  285. Jüppner H, Schipani E, and Silve C. Principles of Bone Biology. Elsevier; 2008:1431-52.
  286. Blomstrand S, Claësson I, and Säve-Söderbergh J. A case of lethal congenital dwarfism with accelerated skeletal maturation. Pediatr Radiol. 1985;15:141-3.
  287. Karaplis AC, Bin He MT, Nguyen A, Young ID, Semeraro D, Ozawa H, et al. Inactivating Mutation in the Human Parathyroid Hormone Receptor Type 1 Gene in Blomstrand Chondrodysplasia. Endocrinology. 1998;139:5255-8.
  288. Karperien M, van der Harten HJ, van Schooten R, Farih-Sips H, den Hollander NS, Kneppers SLJ, et al. A Frame-Shift Mutation in the Type I Parathyroid Hormone (PTH)/PTH-Related Peptide Receptor Causing Blomstrand Lethal Osteochondrodysplasia. The Journal of Clinical Endocrinology &amp; Metabolism. 1999;84(10):3713-20.
  289. Loshkajian A, Roume J, Stanescu V, Delezoide AL, Stampf F, and Maroteaux P. Familial Blomstrand Chondrodysplasia with advanced skeletal maturation:further delineation. Am J Med Genet. 1997;71:283-8.
  290. Zhang P, Jobert AS, Couvineau A, and Silve C. A homozygous inactivating mutation in the parathyroid hormone/parathyroid hormone-related peptide receptor causing Blomstrand chondrodysplasia. J Clin Endocrinol Metab. 1998;83:3365-8.
  291. Hoogendam J, Farih-Sips H, Wynaendts LC, Lowik CW, Wit JM, and Karperien M. Novel mutations in the parathyroid hormone (PTH)/PTH-related peptide receptor type 1 causing Blomstrand osteochondrodysplasia types I and II. J Clin Endocrinol Metab. 2007;92(3):1088-95.
  292. Oostra R, van der Harten J, Rijnders W, Scott R, Young M, and Trump D. Blomstrand osteochondrodysplasia: three novel cases and histological evidence for heterogeneity. Virchows Arch. 2000;436:28-35.
  293. Duchatelet S, Ostergaard E, Cortes D, Lemainque A, and Julier C. Recessive mutations in PTHR1 cause contrasting skeletal dysplasias in Eiken and Blomstrand syndromes. Hum Mol Genet. 2005;14(1):1-5.
  294. Couvineau A, Wouters V, Bertrand G, Rouyer C, Gerard B, Boon LM, et al. PTHR1 mutations associated with Ollier disease result in receptor loss of function. Hum Mol Genet. 2008;17(18):2766-75.
  295. Hopyan S, Gokgoz N, Poon R, Gensure RC, Yu C, Cole WG, et al. A mutant PTH/PTHrP type I receptor in enchondromatosis. Nat Genet. 2002;30(3):306-10.
  296. Rozeman LB, Sangiorgi L, Briaire-de Bruijn IH, Mainil-Varlet P, Bertoni F, Cleton-Jansen AM, et al. Enchondromatosis (Ollier disease, Maffucci syndrome) is not caused by the PTHR1 mutation p.R150C. Hum Mutat. 2004;24(6):466-73.
  297. Collinson M, Leonard SJ, Charlton J, Crolla JA, Silve C, Hall CM, et al. Symmetrical enchondromatosis is associated with duplication of 12p11.23 to 12p11.22 including PTHLH. Am J Med Genet A. 2010;152A(12):3124-8.
  298. Decker E, Stellzig-Eisenhauer A, Fiebig BS, Rau C, Kress W, Saar K, et al. PTHR1 loss-of-function mutations in familial, nonsyndromic primary failure of tooth eruption. Am J Hum Genet. 2008;83(6):781-6.
  299. Frazier-Bowers SA, Hendricks HM, Wright JT, Lee J, Long K, Dibble CF, et al. Novel mutations in PTH1R associated with primary failure of eruption and osteoarthritis. Journal of dental research. 2014;93(2):134-9.
  300. Frazier-Bowers SA, Simmons D, Wright JT, Proffit WR, and Ackerman JL. Primary failure of eruption and PTH1R: the importance of a genetic diagnosis for orthodontic treatment planning. American journal of orthodontics and dentofacial orthopedics : official publication of the American Association of Orthodontists, its constituent societies, and the American Board of Orthodontics. 2010;137(2):160 e1-7; discussion -1.
  301. Risom L, Christoffersen L, Daugaard-Jensen J, Hove HD, Andersen HS, Andresen BS, et al. Identification of six novel PTH1R mutations in families with a history of primary failure of tooth eruption. PLoS One. 2013;8(9):e74601.
  302. Yamaguchi T, Hosomichi K, Narita A, Shirota T, Tomoyasu Y, Maki K, et al. Exome resequencing combined with linkage analysis identifies novel PTH1R variants in primary failure of tooth eruption in Japanese. J Bone Miner Res. 2011;26(7):1655-61.
  303. Konrad M, Schlingmann KP, and Gudermann T. Insights into the molecular nature of magnesium homeostasis. Am J Physiol Renal Physiol. 2004;286(4):F599-605.
  304. Astor MC, Lovas K, Wolff AS, Nedrebo B, Bratland E, Steen-Johnsen J, et al. Hypomagnesemia and functional hypoparathyroidism due to novel mutations in the Mg-channel TRPM6. Endocr Connect. 2015;4(4):215-22.
  305. Janett S, Camozzi P, Peeters GG, Lava SA, Simonetti GD, Goeggel Simonetti B, et al. Hypomagnesemia Induced by Long-Term Treatment with Proton-Pump Inhibitors. Gastroenterol Res Pract. 2015;2015:951768.
  306. Fatuzzo P, Portale G, Scollo V, Zanoli L, and Granata A. Proton pump inhibitors and symptomatic hypomagnesemic hypoparathyroidism. J Nephrol. 2017;30(2):297-301.
  307. Bilezikian JP, Brandi ML, Cusano NE, Mannstadt M, Rejnmark L, Rizzoli R, et al. Management of Hypoparathyroidism: Present and Future. J Clin Endocrinol Metab. 2016;101(6):2313-24.
  308. Brandi ML, Bilezikian JP, Shoback D, Bouillon R, Clarke BL, Thakker RV, et al. Management of Hypoparathyroidism: Summary Statement and Guidelines. J Clin Endocrinol Metab. 2016;101(6):2273-83.
  309. Mittendorf EA, Merlino JI, and McHenry CR. Post-parathyroidectomy hypocalcemia: incidence, risk factors, and management. Am Surg. 2004;70(2):114-9; discussion 9-20.
  310. Khan MI, Waguespack SG, and Hu MI. Medical management of postsurgical hypoparathyroidism. Endocr Pract. 2011;17 Suppl 1:18-25.
  311. Arlt W, Fremerey C, Callies F, Reincke M, Schneider P, Timmermann W, et al. Well-being, mood and calcium homeostasis in patients with hypoparathyroidism receiving standard treatment with calcium and vitamin D. Eur J Endocrinol. 2002;146(2):215-22.
  312. Okano K, Furukawa Y, Morii H, and Fujita T. Comparative Efficacy of Various Vitamin D Metabolites in the Treatment of Various Types of Hypoparathyroidism. The Journal of Clinical Endocrinology &amp; Metabolism. 1982;55(2):238-43.
  313. Astor MC, Løvås K, Debowska A, Eriksen EF, Evang JA, Fossum C, et al. Epidemiology and Health-Related Quality of Life in Hypoparathyroidism in Norway. The Journal of Clinical Endocrinology &amp; Metabolism. 2016;101(8):3045-53.
  314. Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Ahmed SF, et al. Recommendations for Diagnosis and Treatment of Pseudohypoparathyroidism and Related Disorders: An Updated Practical Tool for Physicians and Patients. Hormone research in paediatrics. 2020;93(3):182-96.
  315. Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Usardi A, et al. Diagnosis and management of pseudohypoparathyroidism and related disorders: first international Consensus Statement. Nat Rev Endocrinol. 2018;14(8):476-500.
  316. (FDA) USFaDA. Natpara - Drug Approval Package. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/125511Orig1s000TOC.cfm.
  317. Jolette J, Wilker CE, Smith SY, Doyle N, Hardisty JF, Metcalfe AJ, et al. Defining a Noncarcinogenic Dose of Recombinant Human Parathyroid Hormone 1–84 in a 2-Year Study in Fischer 344 Rats. Toxicologic Pathology. 2006;34(7):929-40.
  318. Capriani C, Irani D, and Bilezikian JP. Safety of osteoanabolic therapy: A decade of experience. Journal of Bone and Mineral Research. 2012;27(12):2419-28.
  319. Krege JH, Gilsenan AW, Komacko JL, and Kellier-Steele N. Teriparatide and Osteosarcoma Risk: History, Science, Elimination of Boxed Warning, and Other Label Updates. JBMR Plus. 2022;6(9):e10665.
  320. Winer KK, Yanovski JA, and Cutler GB, Jr. Synthetic human parathyroid hormone 1-34 vs calcitriol and calcium in the treatment of hypoparathyroidism. Jama. 1996;276(8):631-6.
  321. Winer KK, Ko CW, Reynolds JC, Dowdy K, Keil M, Peterson D, et al. Long-Term Treatment of Hypoparathyroidism: A Randomized Controlled Study Comparing Parathyroid Hormone-(1–34)<i>Versus</i>Calcitriol and Calcium. The Journal of Clinical Endocrinology &amp; Metabolism. 2003;88(9):4214-20.
  322. Winer KK, Sinaii N, Peterson D, Sainz B, Jr., and Cutler GB, Jr. Effects of once versus twice-daily parathyroid hormone 1-34 therapy in children with hypoparathyroidism. The Journal of clinical endocrinology and metabolism. 2008;93(9):3389-95.
  323. Winer KK, Sinaii N, Reynolds J, Peterson D, Dowdy K, and Cutler GB, Jr. Long-term treatment of 12 children with chronic hypoparathyroidism: a randomized trial comparing synthetic human parathyroid hormone 1-34 versus calcitriol and calcium. The Journal of clinical endocrinology and metabolism. 2010;95(6):2680-8.
  324. Winer KK, Yanovski JA, Sarani B, and Cutler Jr GB. A Randomized, Cross-Over Trial of Once-Daily Versus Twice-Daily Parathyroid Hormone 1–34 in Treatment of Hypoparathyroidism. The Journal of Clinical Endocrinology &amp; Metabolism. 1998;83(10):3480-6.
  325. Winer KK, Fulton KA, Albert PS, and Cutler GB, Jr. Effects of pump versus twice-daily injection delivery of synthetic parathyroid hormone 1-34 in children with severe congenital hypoparathyroidism. The Journal of pediatrics. 2014;165(3):556-63.e1.
  326. Winer KK, Zhang B, Shrader JA, Peterson D, Smith M, Albert PS, et al. Synthetic human parathyroid hormone 1-34 replacement therapy: a randomized crossover trial comparing pump versus injections in the treatment of chronic hypoparathyroidism. J Clin Endocrinol Metab. 2012;97(2):391-9.
  327. Santonati A, Palermo A, Maddaloni E, Bosco D, Spada A, Grimaldi F, et al. PTH(1–34) for Surgical Hypoparathyroidism: A Prospective, Open-Label Investigation of Efficacy and Quality of Life. The Journal of Clinical Endocrinology &amp; Metabolism. 2015;100(9):3590-7.
  328. Mittelman SD, Hendy GN, Fefferman RA, Canaff L, Mosesova I, Cole DEC, et al. A Hypocalcemic Child with a Novel Activating Mutation of the Calcium-Sensing Receptor Gene: Successful Treatment with Recombinant Human Parathyroid Hormone. The Journal of Clinical Endocrinology &amp; Metabolism. 2006;91(7):2474-9.
  329. Theman TA, Collins MT, Dempster DW, Zhou H, Reynolds JC, Brahim JS, et al. PTH(1-34) replacement therapy in a child with hypoparathyroidism caused by a sporadic calcium receptor mutation. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2009;24(5):964-73.
  330. Clarke BL, Kay Berg J, Fox J, Cyran JA, and Lagast H. Pharmacokinetics and Pharmacodynamics of Subcutaneous Recombinant Parathyroid Hormone (1–84) in Patients With Hypoparathyroidism: An Open-Label, Single-Dose, Phase I Study. Clinical Therapeutics. 2014;36(5):722-36.
  331. Rubin MR, Sliney J, McMahon DJ, Silverberg SJ, and Bilezikian JP. Therapy of hypoparathyroidism with intact parathyroid hormone. Osteoporosis International. 2010;21(11):1927-34.
  332. Sikjaer T, Amstrup AK, Rolighed L, Kjaer SG, Mosekilde L, and Rejnmark L. PTH(1-84) replacement therapy in hypoparathyroidism: A randomized controlled trial on pharmacokinetic and dynamic effects after 6 months of treatment. Journal of Bone and Mineral Research. 2013;28(10):2232-43.
  333. Cusano NE, Rubin MR, McMahon DJ, Irani D, Anderson L, Levy E, et al. PTH(1-84) is associated with improved quality of life in hypoparathyroidism through 5 years of therapy. The Journal of clinical endocrinology and metabolism. 2014;99(10):3694-9.
  334. Cusano NE, Rubin MR, McMahon DJ, Zhang C, Ives R, Tulley A, et al. Therapy of Hypoparathyroidism with PTH(1–84): A Prospective Four-Year Investigation of Efficacy and Safety. The Journal of Clinical Endocrinology &amp; Metabolism. 2013;98(1):137-44.
  335. Sikjaer T, Rejnmark L, Rolighed L, Heickendorff L, and Mosekilde L. The effect of adding PTH(1-84) to conventional treatment of hypoparathyroidism: A randomized, placebo-controlled study. Journal of Bone and Mineral Research. 2011;26(10):2358-70.
  336. Rubin MR, Dempster DW, Sliney J, Jr., Zhou H, Nickolas TL, Stein EM, et al. PTH(1-84) administration reverses abnormal bone-remodeling dynamics and structure in hypoparathyroidism. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2011;26(11):2727-36.
  337. Cusano NE, Rubin MR, McMahon DJ, Irani D, Tulley A, Sliney J, Jr., et al. The effect of PTH(1-84) on quality of life in hypoparathyroidism. The Journal of clinical endocrinology and metabolism. 2013;98(6):2356-61.
  338. Mannstadt M, Clarke BL, Vokes T, Brandi ML, Ranganath L, Fraser WD, et al. Efficacy and safety of recombinant human parathyroid hormone (1–84) in hypoparathyroidism (REPLACE): a double-blind, placebo-controlled, randomised, phase 3 study. The Lancet Diabetes &amp; Endocrinology. 2013;1(4):275-83.
  339. Clarke BL, Vokes TJ, Bilezikian JP, Shoback DM, Lagast H, and Mannstadt M. Effects of parathyroid hormone rhPTH(1-84) on phosphate homeostasis and vitamin D metabolism in hypoparathyroidism: REPLACE phase 3 study. Endocrine. 2017;55(1):273-82.
  340. Khan AA, Rubin MR, Schwarz P, Vokes T, Shoback DM, Gagnon C, et al. Efficacy and Safety of Parathyroid Hormone Replacement With TransCon PTH in Hypoparathyroidism: 26-Week Results From the Phase 3 PaTHway Trial. J Bone Miner Res. 2023;38(1):14-25.
  341. Nemeth EF, and Goodman WG. Calcimimetic and Calcilytic Drugs: Feats, Flops, and Futures. Calcified tissue international. 2015;98(4):341-58.
  342. Dong B, Endo I, Ohnishi Y, Kondo T, Hasegawa T, Amizuka N, et al. Calcilytic Ameliorates Abnormalities of Mutant Calcium-Sensing Receptor (CaSR) Knock-In Mice Mimicking Autosomal Dominant Hypocalcemia (ADH). Journal of Bone and Mineral Research. 2015;30(11):1980-93.
  343. Hannan FM, Walls GV, Babinsky VN, Nesbit MA, Kallay E, Hough TA, et al. The Calcilytic Agent NPS 2143 Rectifies Hypocalcemia in a Mouse Model With an Activating Calcium-Sensing Receptor (CaSR) Mutation: Relevance to Autosomal Dominant Hypocalcemia Type 1 (ADH1). Endocrinology. 2015;156(9):3114-21.
  344. Roberts MS, Gafni RI, Brillante B, Guthrie LC, Streit J, Gash D, et al. Treatment of Autosomal Dominant Hypocalcemia Type 1 With the Calcilytic NPSP795 (SHP635). Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2019;34(9):1609-18.
  345. Hu J, and Spiegel AM. Structure and function of the human calcium-sensing receptor: insights from natural and engineered mutations and allosteric modulators. J Cell Mol Med. 2007;11(5):908-22.
  346. Letz S, Haag C, Schulze E, Frank-Raue K, Raue F, Hofner B, et al. Amino alcohol- (NPS-2143) and quinazolinone-derived calcilytics (ATF936 and AXT914) differentially mitigate excessive signalling of calcium-sensing receptor mutants causing Bartter syndrome Type 5 and autosomal dominant hypocalcemia. PLoS One. 2014;9(12):e115178.
  347. Letz S, Rus R, Haag C, Dörr H-Gn, Schnabel D, Möhlig M, et al. Novel Activating Mutations of the Calcium-Sensing Receptor: The Calcilytic NPS-2143 Mitigates Excessive Signal Transduction of Mutant Receptors. The Journal of Clinical Endocrinology &amp; Metabolism. 2010;95(10):E229-E33.
  348. Roszko KL, Bi RD, and Mannstadt M. Autosomal Dominant Hypocalcemia (Hypoparathyroidism) Types 1 and 2. Front Physiol. 2016;7:458.
  349. Hannan FM, Babinsky VN, and Thakker RV. Disorders of the calcium-sensing receptor and partner proteins: insights into the molecular basis of calcium homeostasis. Journal of molecular endocrinology. 2016;57(3):R127-R42.
  350. Babinsky VN, Hannan FM, Gorvin CM, Howles SA, Nesbit MA, Rust N, et al. Allosteric Modulation of the Calcium-sensing Receptor Rectifies Signaling Abnormalities Associated with G-protein α-11 Mutations Causing Hypercalcemic and Hypocalcemic Disorders. The Journal of biological chemistry. 2016;291(20):10876-85.
  351. Roszko KL, Bi R, Gorvin CM, Brauner-Osborne H, Xiong XF, Inoue A, et al. Knockin mouse with mutant Galpha(11) mimics human inherited hypocalcemia and is rescued by pharmacologic inhibitors. JCI Insight. 2017;2(3):e91079.
  352. Xiong X-F, Zhang H, Underwood CR, Harpsøe K, Gardella TJ, Wöldike MF, et al. Total synthesis and structure-activity relationship studies of a series of selective G protein inhibitors. Nat Chem. 2016;8(11):1035-41.
  353. Sato K, Hasegawa Y, Nakae J, Nanao K, Takahashi I, Tajima T, et al. Hydrochlorothiazide Effectively Reduces Urinary Calcium Excretion in Two Japanese Patients with Gain-of-Function Mutations of the Calcium-Sensing Receptor Gene. The Journal of Clinical Endocrinology &amp; Metabolism. 2002;87(7):3068-73.
  354. Mantovani G, Ferrante E, Giavoli C, Linglart A, Cappa M, Cisternino M, et al. Recombinant human GH replacement therapy in children with pseudohypoparathyroidism type Ia: first study on the effect on growth. J Clin Endocrinol Metab. 2010;95(11):5011-7.
  355. Ertl DA, de Nanclares GP, Juppner H, Hanna P, Pagnano A, Pereda A, et al. Recombinant growth hormone improves growth and adult height in patients with maternal inactivating GNAS mutations. Eur J Endocrinol. 2023;189(1):123-31.

 -- In memory of Dr. Geoffrey N. Hendy --

 

Dietary Advice for Individuals with Diabetes

ABSTRACT

 

The chapter summarizes the current information available from a variety of scientifically based guidelines and resources on dietary advice for those with diabetes. It is a practical overview for health care practitioners working in diabetes management. The chapter is divided into sections by content and includes sources for further reading. A primary message is that nutrition plans should meet the specific needs of the patient and take into consideration their ability to implement change. Often starting with small achievable changes is best, with larger changes discussed as rapport builds. Referral to medical nutrition therapy (MNT) provided by a Registered Dietitian Nutritionist (RDN) and a diabetes self- management education and support (DSMES) program is highlighted.

 

INTRODUCTION

 

This chapter will summarize current information available from a variety of evidence-based guidelines and resources on dietary advice for those with diabetes. The modern diet for those with diabetes is based on concepts from clinical research, portion control, and individualized lifestyle change. It requires open and honest communication between health care practitioner and patient and cannot be delivered by giving a person a diet sheet in a one-size-fits- all approach. The lifestyle modification guidance and support needed most often requires a team effort, ideally including a registered dietitian(RD) or registered dietitian nutritionist (RDN), or a referral to a diabetes self- management education and support (DSMES) program that includes dietary advice. Current (2024) recommendations of the American Diabetes Association (ADA) promote all health care professionals to refer people with diabetes for individualized medical nutrition therapy (MNT) provided by an RDN at diagnosis and as needed throughout the life span, in addition to DSMES (1). It is very important to note that dietary recommendations for those with diabetes are virtually the same recommendations for diabetes prevention and the health of the general population; however, it cannot be excluded that people with diabetes will require additional support to meet the recommendations.

 

Fang et al, reported that although there has been continued improvements in risk factor control and adherence to preventative practices over the past decades, half of U.S. adults with diabetes do not meet the recommended goals for diabetes care in 2015-2018 (2). This is a current and ongoing issue. Diet and lifestyle recommendations are cornerstones of advice to prevent and manage diabetes, however there are recognized barriers to heeding advice and implementing lifestyle change. First, there is a plethora of dietary information for diabetes management available from many sources, although not all is evidence-based or current. There are also social, cultural, and personal preferences unique to each individual that must be taken into consideration when making long-term dietary change. Many health care practitioners are not adequately trained to be confident in delivering dietary advice, and many food environments do not support healthy dietary intakes for all. There are also commercial determinants of health that influence dietary intakes, such as marketing advertising, and price discounting on certain foods. The following recommendations come from evidence-based guideline development processes and emphasize practical suggestions for implementing dietary advice for most individuals with diabetes.

 

GENERAL GOALS

 

Dietary advice for those with diabetes has evolved and have become more flexible and patient centered over time. Nutrition goals from the American Diabetes Association (ADA) 2024 include the following: (1)

 

  1. To promote and support healthful eating patterns, emphasizing a variety of nutrient-dense foods in appropriate portion sizes, to improve overall health and:
    • achieve and maintain body weight goals.
    • attain individualized glycemic, blood pressure, and lipid goals.
    • delay or prevent the complications of diabetes.
  2. To address individual nutrition needs based on personal and cultural preferences, health literacy and numeracy, access to healthful foods, willingness and ability to make behavioral changes, and existing barriers to change.
  3. To maintain the pleasure of eating by providing nonjudgmental messages about food choices while limiting food choices only when indicated by scientific evidence.
  4. To provide an individual with diabetes the practical tools for developing healthy eating patterns rather than focusing on individual macronutrients, micronutrients, or single foods.

 

The American Association of Clinical Endocrinologists (AACE) guidelines have similar nutrition goals for people with type2 diabetes (3).

 

Putting Goals Into Practice

 

How should these goals best be put into practice? The following guidelines summarized from the ADA Standards of Care will address the above goals and provide guidance on nutrition therapy based on numerous scientific resources. The Diabetes Control and Complications Trial (DCCT) and other studies demonstrated the added value individualized consultation with a registered dietitian familiar with diabetes treatments, along with regular follow-up, has on long-term outcomes and is highly recommended to aid in lifestyle compliance (4). Medical nutrition therapy (MNT) implemented by a registered dietitian is associated with A1C reductions of 1.0–1.9% for people with type 1 diabetes and 0.3–2.0% for people with type 2 diabetes (1).

 

Target Guidelines For Macronutrients: The 3 Major Components Of Diet

 

Many studies have been completed to attempt to determine the optimal combination of macronutrients. Based on available data, the best mix of carbohydrate, protein, and fat depends on the individual metabolic goals and preferences of the person with diabetes. It’s most important to ensure that total energy intake is kept in mind for weight loss or maintenance (1).

 

CARBOHYDRATES

 

The primary goal in the management of diabetes is to achieve as near normal regulation of blood glucose as possible. Both the type and total amount of carbohydrate (CHO) consumed influences glycemia. Carbohydrate intake should emphasize nutrient-dense carbohydrate sources that are high in fiber (at least 14 g fiber per 1,000 kcal) and minimally processed (1). Dietary carbohydrate includes sugars, starch, and dietary fiber. Higher intakes of sugars are associated with weight gain and greater incidence of dental caries (5). Conversely, higher intakes of dietary fiber are associated with reduced non-communicable disease and premature mortality occurrence as well as improvements in body weight, cholesterol concentrations, and blood pressure (6, 7). These benefits with higher fiber intakes have been observed in the general population, for those with type 1, type 2, and pre diabetes, (8) and those with hypertension or heart disease (9).With this guidance in mind, eating plans should emphasize non-starchy vegetables, fruits, legumes, and whole grains, as well as dairy products with minimal added sugars (1, 10). There is less consistency of evidence for recommending an amount of overall CHO in the diet (1). This is in line with current World Health Organization for carbohydrate intakes for adults and children which stress the type of carbohydrate is important, with recommendations for fiber and vegetable and fruit intake, but no recommendations on CHO amount (7). Recent dietary guidelines for diabetes management from the European Association for the Study of Diabetes stress that a wide range of carbohydrate intakes can be appropriate, however both very high (>70%Total Energy (TE)) and low (<40%TE) intakes are associated with premature mortality (10). A recent comprehensive Cochrane systematic review of randomized controlled trials (RCTs) of adults with overweight or obesity with or without type 2 diabetes concluded that there is probably little to no difference in weight reduction and changes in cardiovascular risk factors up to two years' follow-up, when overweight and obese participants without and with T2DM are randomized to either low-carbohydrate or balanced-carbohydrate weight-reducing diets (11).Some of the reasons for these findings of a lack of effect with lower carbohydrate diets may be that: interventions do not consider the type of carbohydrate being consumed, with dietary fiber and sugar having differing physiological effects; the differing definitions of low CHO diets being applied; what CHO is replaced with; and that diets lower in CHO maybe difficult to maintain in the long term as they are not consistent with the socio, cultural, and personal preference of many. Current ADA recommendations relating to CHO are: (1)

 

  • Emphasize minimally processed, nutrient-dense, high-fiber sources of carbohydrate (at least 14 g fiber per 1,000 kcal).
  • People with diabetes and those at risk are advised to replace sugar-sweetened beverages (including fruit juices) with water or low-calorie or no-calorie beverages as much as possible to manage glycemia and reduce risk for cardiometabolic disease and minimize consumption of foods with added sugar that have the capacity to displace healthier, more nutrient-dense food choices.
  • Provide education on the glycemic impact of carbohydrate, fat, and protein tailored to an individual’s needs, insulin plan, and preferences to optimize mealtime insulin dosing.
  • When using fixed insulin doses, individuals should be provided with education about consistent patterns of carbohydrate intake with respect to time and amount while considering the insulin action time, as it can result in improved glycemia and reduce the risk for hypoglycemia.

 

Dietary Fiber

 

Current recommendations from the American Diabetes Association are that adults with diabetes should consume high fiber foods (at least 14g fiber per 1,000 kcal) (1). Current recommendations from the European Association for the Study of Diabetes are that adults with diabetes should consume at least 35g dietary fiber per day (or 16.7g per 1,000 kcal) (10).These two values are aligned, and higher than current World Health Organization recommendations for the general population of at least 25g dietary fiber per day, (7) although all three recommendations recognize a minimum intake level, with greater benefits observed with higher intakes. These values are appreciably higher than current dietary fiber intakes in the United States, which is approximately 16g per day. Our understanding of the importance of dietary fiber has changed in recent years. Dietary fiber is carbohydrate that is not digested by the stomach or absorbed in the GI tract. Instead, it is either degraded in the colon by the gut microbiota, or passes through the human body intact. Higher intakes of dietary fiber are associated with lower all-cause mortality, heart disease, T2 diabetes incidence, and certain cancers such colorectal cancer when compared with lower fiber intakes (6). The benefits for childhood intakes of dietary fiber and health outcomes later in life remain uncertain (12). There are several established physiological pathways that might explain these associations, such as reducing postprandial glycemia, competitive inhibition of saturated fat in the small intestine, and greater satiety leading to reduce subsequent intake. There are also more novel pathways proposed, such as modulation of the gut microbiota to increase branched and short chain fatty acids. Current recommendations by the World Health Organization are to obtain “naturally occurring dietary fiber as consumed in food” (7). Fiber supplements however are used frequently as additional dietary fiber sources, and may help individuals reach their fiber recommendations when sufficient amounts cannot be obtained from food alone. Fiber supplements can be extracted fiber (taken from a plant source) or synthetic. Few fiber supplements have been studied for physiological effectiveness to the same degree as inherent dietary fiber, so current best advice is to consume foods that are high in fiber (1, 7, 13).Recommended food sources of dietary fiber are minimally processed whole grains, vegetables, whole fruit and legumes (1, 7).  An emphasis on minimally processed is made, as processing may reduce the benefits associated with intakes of these foods, (14-16) as well as introduce added nutrients such as saturated fats, sodium, and added sugars.

 

The website below contains links to a comprehensive table listing fiber content of foods, and a calculator to help select foods with higher fiber content to help reach daily fiber goals.

http://www.webmd.com/diet/healthtool-fiber-meter. In the Endotext chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” in the Lipid and Lipoprotein section provides several tables providing information on the fiber content of various foods.

 

Starch

 

Starch comprises most of the carbohydrates consumed globally, and is the storage carbohydrate found in refined cereals, potatoes, legumes, and bananas (16). Starch comprises two polymers: amylose (DP ~ 103) and amylopectin (DP ~ 104–105). Most cereal starches comprise 15–30% amylose and 70–85% amylopectin. In their raw form, most starches are resistant to digestion by pancreatic amylase, but gelatinize in heat and water, permitting rapid digestion (16). Dietary starch intake is rarely directly reported, so the health effects of dietary starch intake are often assessed through key sources, such as refined grains and potatoes. For potatoes, meta-analyses of prospective observational studies have identified the health effects are largely determined by the cooking method (17). Fried and salted potatoes were associated with higher incidence of type 2 diabetes and hypertension. Boiled and roasted potatoes were not associated with increased or decreased risk to health (17). Some starches escape digestion, either naturally or due to food processing; these starches are called resistant starches.

 

Resistant Starches

 

Resistant starches are starch enclosed within intact cell walls. These include some legumes, starch granules in raw potato, retrograde amylose from plants modified to increase amylose content, or high-amylose containing foods, such as specially formulated cornstarch, which are not digested and absorbed as glucose. Resistant starches avoid digestion in the small intestine so do not contribute to postprandial glycemia and diabetes risk, and are instead fermented in the colon by the microbiota.

 

Sugars (Nutritive Sweeteners)

 

Sucrose, also known as “table sugar,” is a disaccharide composed of one glucose and one fructose molecule and provides 4 kcals per gram (16). Available evidence from clinical studies does not indicate that the overall amount of dietary sucrose is related to type 2 diabetes incidence, however it is related to body weight gain and increased dental caries (5). Given the association between excess body weight and type 2 diabetes occurrence, (18) there is rationale to promote a reduction of sugar intake related to diabetes occurrence, and replace sugar-sweetened beverages (including fruit juices) with water or no/low calorie beverages as much as possible (1).

 

Fructose is a naturally occurring monosaccharide found in fruits, some vegetables, and honey. High fructose corn syrup is used abundantly within the United States in processed foods as a less expensive alternative to sucrose. Fructose consumed in naturally occurring in foods such as fruit, (that also contain fiber) may result in better glycemic control compared with isocaloric intake of sucrose or fructose added to food, and is not likely to have detrimental effects on triglycerides as long as intake is not excessive (<12% energy).

 

A meta-analysis of 18 controlled feeding trials in people with diabetes compared the impact of fructose with other sources of carbohydrate on glycemic control. The analysis found that an isocaloric exchange of fructose for carbohydrates did not significantly affect fasting glucose or insulin and reduced glycated blood proteins in these trials of less than 12 weeks duration. The short duration is a potential limitation of the studies (19).  Evidence exists that consuming high levels of fructose-containing beverages may have particularly adverse effects on selective deposition of ectopic and visceral fat, lipid metabolism, blood pressure, and insulin sensitivity compared with glucose-sweetened beverages (20). Thus, recommendations for dietary fructose tend to promote the reduction of fructose added to food, such as in fructose-containing beverages, while promoting whole fruit which can contain intrinsic fructose.

 

Non-Nutritive Sweeteners

 

Non-nutritive sweeteners provide insignificant amounts of energy and elicit a sweet sensation without increasing blood glucose or insulin concentrations. There are several FDA-approved sweeteners found to be safe when consumed within FDA acceptable daily intake amounts (ADI) (Table 1) (21).

 

Table 1. NON-NUTRITIVE SWEETENERS

Name

Main Source

Sucralose (Splenda®)

Sucralose is synthesized from regular sucrose, but altered such that it is not absorbed. Sucralose is 600 times sweeter than sucrose. It is heat stable and can be used in cooking. It was approved for use by the FDA in 1999.

Saccharine

(Sugar Twin®, Sweet ‘N Low®)

Saccharine is 200 to 700 times sweeter than sugar. A cancer-related warning label was removed in 2000 after the FDA determined that it was generally safe.

Acesulfame K (Ace K, Sunette)

Acesulfame is 200 times sweeter than sucrose. It can be used in cooking. The bitter aftertaste of acesulfame can be greatly decreased or eliminated by combining acesulfame with another sweetener.

Neotame

Neotame is a derivative of the dipeptide phenylalanine and aspartic acid. It is 7,000-13,000 times sweeter than sucrose and does not have a significant effect on fasting glucose or insulin levels in persons with type 2 diabetes.

Aspartame (Equal®,NutraSweet®)

Aspartame is a methyl ester of aspartic acid and phenylalanine dipeptide. Although aspartame provides 4 kcal/g, the intensity of the sweet taste(200x sweeter than sucrose) means that very small amounts are required. The FDA requires any foods containing aspartame to have an informational label statement: “Phenylketonurics: contains phenylalanine.” Patients with phenylketonuria should avoid products containing Aspartame. Controversy has existed for many years around safety of this sweetener, but not from any major organizations.

Stevia (Truvia®)

Stevia derived from the plant stevia rebaudiana, is a non-caloric, natural sweetener. Stevia has been used as a sweetener and as a medicinal herb since ancient times and appears to be well-tolerated. It has an intensely sweet taste.

Luo han guo

Luo han guo is also known as monk fruit, or Swingle fruit extract. It is 150- 300 times sweeter than sucrose, and may have an aftertaste at high levels.

 

A review of 29 RCTs which included 741 people, 69 of which have type 2 diabetes, indicated that artificial sweeteners on their own do not raise blood glucose levels, but the content of the food or drink containing the artificial sweetener must be considered, especially for those with diabetes (22). This sentiment was echoed in recent WHO guidance on non-nutritive sweeteners for the general population (23) where their use was not recommended for weight loss, as the overall content of the processed food or drink was important.

 

Practical Tips For Carbohydrate Intake

 

  • Base meals and snacks around high fiber foods, such as whole grains, vegetables, whole fruit, and legumes.
  • Common whole grains include whole wheat, whole oats, brown rice, barley, and quinoa.
  • When purchasing wholegrain foods, check the label to make sure that the wholegrain is the first ingredient listed, and that energy from sugars is <10%.
  • Consume fruit, but chose whole fruit over dried, juiced, or further processed fruit.
  • Legumes are an excellent and cheap source of fiber and protein. Replace ground meat in meals such as casserole with lentils or legumes.
  • Strive to include a variety of vegetables in your meals each day, avoiding deep fried and heavily salted options.

 

FAT

 

Evidence is inconclusive for an ideal amount of total fat intake for people with diabetes; therefore, goals should be individualized.

 

In line with advice for the general public, people with diabetes should look to replace saturated and trans fats in the diet with mono and poly unsaturated fats (24). This is principally to lessen the increased risk of cardiovascular disease with high saturated and trans-fat intakes. Recent meta-analyses have found that decreasing the amount of saturated fatty acids and trans fatty acids, the principal dietary fatty acids linked to elevating LDL cholesterol, reduces the risk of CVD(25). The World Health Organization and American College of Cardiology currently recommend limiting the amount of dietary saturated and trans-fat intake (24, 26). Recommendations from the Institute of Medicine and the Academy of Nutrition and Dietetics for healthy individuals are that 20% to 35% of total energy should come from fat (27).Recommendations to reduce total fat intake are largely due to the high energy content of dietary fats, more so than protein or carbohydrate, and the risks associated with higher saturated fat intakes. Current recommendations for fat intakes from the American Diabetes Association focus on fat quality and its sources rather than quantity (1). They recommend:

 

  • Counsel people with diabetes to consider an eating plan emphasizing elements of a Mediterranean eating pattern, which is rich in monounsaturated and polyunsaturated fats and long-chain fatty acids such as fatty fish, nuts, and seeds, to reduce cardiovascular disease risk and improve glucose metabolism.

 

The American Heart Association has developed the Fat Facts to help individuals learn more about healthy vs. unhealthy fats. Among the campaign's top priorities is to encourage replacing high trans-fat partially hydrogenated vegetable oils,animal fats, and tropical oils with healthier oils and foods higher in unsaturated fats — monounsaturated and polyunsaturated.

 

See more at: https://www.heart.org/en/healthy-living/healthy-eating/eat-smart/fats/the-facts-on-fats

 

Monounsaturated Fatty Acids

 

Monounsaturated fats (MUFA) are in foods such as avocado, some fish, nuts, and nut butters. MUFA are also found in vegetable oils such as olive, peanut, avocado, and canola oil. Several large prospective observational studies have documented that diets rich in MUFA or PUFA and lower in saturated fat are associated with a reduced risk of CVD (28).Meta-analysis of RCTs comparing diets higher in MUFA vs CHO or PUFA demonstrated that high MUFA containing diets can improve metabolic parameters and reduce cardiovascular disease risk in people with T2D (29, 30).

 

Polyunsaturated Fatty Acids

 

Polyunsaturated fats (PUFAs) are found in foods such as walnuts, sunflower seeds, and some fish such as salmon, mackerel, herring, and trout. PUFA are also found in vegetable oils such as corn oil, safflower oil, and soybean oil. Both PUFA and MUFA are usually liquid at room temperature. A meta-analysis of feeding trials has indicated consistent positive effects when other macronutrients, such as saturated fats, are replaced with PUFA on glycemia, insulin resistance, and insulin secretion capacity (31). Substitution data from prospective observational studies also indicates that replacing saturated and trans fats with PUFA reduces all-cause mortality and coronary heart disease, (25) with a smaller body of evidence in those with diabetes indicating similar improvements in cardiovascular disease risk (30).

 

A few specific types of PUFA are referred to as Omega-3 fats. These are called eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and alpha-linolenic acid (ALA). These fats are particularly singled out and recommended to prevent or treat CVD; however, evidence does not support a beneficial role for the routine use of n-3 dietary supplements in diabetes management (1) or for the general population. EPA and DHA are found in fatty fish. ALA is found in nuts and seeds. Studies on the effect of omega-3 fatty acids (both from food and supplements) in persons with diabetes are limited and have been inconclusive (20). In addition to providing EPA and DHA, regular fish consumption may help reduce triglycerides by replacing other foods higher in saturated and trans fats from the diet, such as fatty meats and full-fat dairy products. Preparing fish without frying or adding cream-based sauces is recommended. Fish with high amounts of EPA and DHA include salmon, albacore tuna, mackerel, sardines, herring, and lake trout. Nuts and seeds high in ALA include walnuts, flax seeds, chia seeds and soybeans (16).

 

Saturated Fats

 

Saturated fats are usually solid or almost solid at room temperature. All land animal fats, such as those in meat, poultry, and dairy products, are predominantly saturated. Processed and fast foods also contain high amounts of saturated fats.Some vegetable oils also can be saturated, including palm, palm kernel, and coconut oils (16). Oil such as coconut and palm (sometimes referred to as tropical oils) are touted as healthful saturated fats since they are derived from plants, however this is not accurate (25). Current ADA recommendations are to limit all sources of saturated fats (1). The World Health Organization recommends limited consumption of saturated fats to less than 10% of total energy intake, (24)which is far less than the current average intake. When cooking with oil, choose non-tropical vegetable oils such as canola, corn, olive, peanut, safflower, soybean, and sunflower oils (16).

 

Few research studies have been undertaken to look at the difference between the amount of saturated fatty acids (SFA) in the diet and glycemic control and CVD risk in people with diabetes (30). The ADA recommends people with diabetes follow the guidelines for the general population (20).

 

In general, saturated fats are discouraged because they increase LDL-cholesterol and total cholesterol concentrations (24). Diets high in saturated fats have been implicated in an increased risk of cardiovascular disease. Three RCTs found that diets containing ≤7% SFA and ≤200 mg/day cholesterol reduced LDL cholesterol level from 9% to 12% compared tobaseline values or to a more standard Western-type diet (32). As saturated fats are progressively decreased in the diet,they should be replaced with unsaturated fats and high fiber carbohydrates, and not with trans fats or refined carbohydrates (25).

 

Trans Fats

 

Trans fatty acids (TFA) are also called hydrogenated fats, which are fats created when oils are "partially hydrogenated" (16). The process of hydrogenation changes the chemical structure of unsaturated fats by adding hydrogen atoms, or “saturating” the fat. Hydrogenation converts liquid oil into stick margarine or shortening. Manufacturers use hydrogenation to increase product stability and shelf-life. A large quantity of these fats can be produced at one time, saving manufacturing costs. Research trials indicate that TFA can increase LDL cholesterol and lower HDL cholesterol (33). Meta-analysis of prospective observational studies indicate higher intakes of trans fats are associated with higher cases of all-cause mortality, cardiovascular disease, and coronary heart disease (25). Although less prevalent by volume in the food supply, trans fats appear at least as harmful to health as saturated fats (25). Due to the observations from both RCTs and prospective observational studies, the World Health Organization currently recommends that the total intake of trans fats be less than 1% of total energy intake (24). With the mandatory TFA labeling in the United States in 2006, a big push has been made by food manufacturers to remove TFA from processed and baked goods. Although the TFA content in foods has decreased recently (through food reformulation), it is important to monitor the type of fat used to replace TFA, as it might be saturated fat. The FDA has determined that trans fats are no longer considered generally recognized as safe (GRAS).While manufacturers cannot add TFAs to foods anymore, they may still be produced during the food manufacturing, so consumers should still check the nutrition information panel on foods. The main sources of trans fats in the food supply today are highly processed foods such as cakes, cookies, potato chips, and animal products. They can also be produced in the home when frying foods in fat at high temperatures. While most trans fats in the diet now are created during food manufacturing, smaller amounts of trans fats are also found in ruminant animals (cows and sheep). At present there is insufficient evidence to indicate that the health effects differ between trans fats that are created in food manufacturing or ruminant derived, (25) so advice to reduce trans-fat intakes relates to total trans-fat (24).

 

Cholesterol

 

The body makes enough cholesterol for physiological functions, so it is not needed through foods. Older dietary guidelines formerly recommended avoiding or limiting consumption of foods high in cholesterol, in the idea that their intake would raise our own circulating cholesterol levels. Now however, it is understood that saturated fat intake has a stronger influence on human cholesterol levels, so recommendations focus on reducing saturated fat as the priority (24).

 

Table 2. DIETARY FATS

Type of Fat

Main Source

Monounsaturated

Canola, peanut, and olive oils; avocados; nuts such as almonds, hazelnuts, and pecans; and seeds such as pumpkin and sesame seeds.

Polyunsaturated

Sunflower, corn, soybean, and flaxseed oils, and also in foods such as walnuts, flax seeds, and fish.

Saturated

Whole milk, butter, cheese, and ice cream; red meat; chocolate; coconuts, coconut milk, coconut oil and palm oil.

Trans

Some margarines; vegetable shortening; partially hydrogenated vegetable oil; deep-fried foods; many fast foods; some commercial baked goods (check labels).

 

Stanols And Sterols

 

Plant sterols are naturally occurring cholesterol derivatives from vegetable oils, nuts, corn, woods, and beans. Hydrogenation of sterols produces stanols. The generic term to describe both sterols, stanols, and their esters is phytosterols. An important role of phytosterols is their ability to block absorption of dietary and biliary cholesterol from the gastrointestinal tract. The LDL lowering property of both sterols and stanols is considered equivalent in short term studies(34). The amounts of sterols and stanol esters found naturally in a normal diet are insufficient to have a therapeutic effect. Thus, many manufacturers add them to various foods for their LDL cholesterol lowering effects. You can find added phytosterols in margarine spreads, juices, yogurts, cereals, and even granola bars. Individuals with diabetes and dyslipidemia may be able to reduce total and LDL cholesterol by consuming at least 2 grams per day of plant stanols or sterols found in enriched foods (20). The evidence on long term use and in people with diabetes is less substantiated, as not many studies have been completed (35).

 

Practical Tips On Fat Intake

 

  • Fat intake should come primarily from good sources of mono and polyunsaturated fats: nuts and seeds, avocados, fish, and oils such as olive, canola, soybean, sunflower, and corn.
  • Limit intake of saturated fats by cutting back on processed and fast foods, red meat, and full-fat dairy foods. Try replacing red meat with beans, nuts, skinless poultry, and fish whenever possible, and switching from whole milk and other full-fat dairy foods to lower fat
  • In place of butter or margarine, use liquid vegetable oils rich in polyunsaturated and monounsaturated fats in cooking and at the table.
  • Keep trans-fat intakes as low as possible. Check food labels for trans fats, and limit fried

 

PROTEIN

 

Protein intake goals should be individualized based on an individual’s current eating patterns. The ADA Standards of Medical Care in Diabetes-2024 state that there is no evidence that adjusting the daily level of protein intake (typically1–1.5g/kg body weight/day or 10–20% total energy) will improve health in individuals without diabetic kidney disease, and research is inconclusive regarding the ideal amount of dietary protein to optimize either glycemic control or cardiovascular disease (CVD) risk (1). Some research has found successful management of weight and type 2 diabetes with meal plans including slightly higher levels of protein (23–32% total energy) for periods up to one year for those without kidney disease (10). Those with diabetic kidney disease (with albuminuria and/or reduced estimated glomerular filtration rate) should aim to maintain dietary protein at the recommended daily allowance of no more than 0.8g/kg desirable body weight/day (or 10-15% total energy) (10). The National Kidney Foundation recommends 0.8 g protein/kg desirable body weight for people with diabetes and stages 1–4 chronic kidney disease as a means of reducing albuminuria and stabilizing kidney function (36). Reducing the amount of dietary protein below 10% total energy is not recommended as it places people at risk of protein inadequacy (10).

 

The ADA recommends that in individuals with type 2 diabetes, ingested protein can increase insulin response without increasing plasma glucose concentrations. Therefore, carbohydrate sources high in protein should not be used to treat or prevent hypoglycemia (1). Further research is required to identify if the dietary source of protein (animal or plant) is important for health and diabetes. There is emerging evidence to suggest that plant sourced proteins may be superior for health, (1) however it is not yet known if this is due to the amino acid compositions of the proteins or unadjusted effects from the accompanying nutrients, such as saturated fats in meat sources and dietary fiber in plant sources of protein. Replacement of red meat in the diet with plant-based protein sources (such as beans and legumes) appears to produce both health and environmental co-benefits, as well as being cheaper (37-39).

 

Practical Tips For Protein Intake

 

  • Ideal plant protein sources include legumes, lentils, tofu, and tempeh (1/2c = 2 oz protein). Plant-based meat alternatives maybe also be used (i.e. Quorn), but be wary of meat alternatives that have high sodium and saturated fat
  • Nuts or seeds are another plant-based protein source to be encouraged, such as almonds, cashews, hazelnuts, filberts, Brazil nuts, macadamias, peanuts, pecans, walnuts, or sunflower, pumpkin seed or linseed. Nut butters are also a plant-based protein source but be mindful of added sodium and sugars.
  • Good sources of lean animal protein include: skinless poultry, lower fat cuts of beef or pork, fish or egg, and reduced fat dairy products (i.e. low fat or skim milk/yogurt, and cheese).
  • Protein sources should be a supplement to vegetables, fruits and whole grains for most meals, and not the entire meal.

 

TARGET GUIDELINES FOR MICRONUTRIENTS

 

There is no clear evidence that dietary supplementation with vitamins (such as Vitamin D), minerals (such as chromium), herbs, or spices (such as cinnamon or aloe vera) can improve outcomes in diabetes management where there are no underlying deficiencies. There is insufficient evidence for dietary supplements to be recommended for the purposes of improving glycemic control (1).

 

People with diabetes should be aware of the necessity for meeting vitamin and mineral needs from natural food sources through intake of a balanced diet. Specific populations, such as older adults, pregnant or lactating women, strict vegetarians or vegans, and individuals on very low energy diets may benefit from a multivitamin mineral supplement (1).Excessive doses of certain vitamin or mineral supplements when there is no deficiency has been shown to be of no benefit and may even be harmful. There is some evidence that those on metformin therapy are at higher risk of B12 deficiency and may need Vitamin B12 supplementation if tests indicate a deficiency (1, 40).

 

VITAMINS

 

Since type 2 diabetes is a state of increased oxidative stress, interest in recommending large doses of antioxidant vitamins has been high. Current studies demonstrate no benefit of carotene and Vitamins E and C in respect to improved glycemic control or treatment of complications. Routinely supplementing the diet with antioxidant supplements is not recommended due to lack of evidence showing benefit in large, placebo-controlled clinical trials and concerns regarding potential long-term safety (1, 40). There is also not adequate evidence to recommend routine Vitamin D supplementation without deficiency (1, 41).

 

MINERALS

 

Sodium

 

As for the general population, those with diabetes should limit sodium consumption to 2,300 mg/day (20, 42). Active steps to reduce current sodium intakes is necessary, as current intakes in the United States are around 3,400 mg/day, nearly 50% more than the recommended limit. The majority of sodium consumed is from processed foods. Food manufacturers and restaurants will need to provide additional reduced sodium alternatives to help accomplish consumption targets. For those with diabetes and hypertension, additional lifestyle modification beyond reducing sodium intake can be helpful, including: loss of excess body weight; increasing consumption of vegetables and fruit (8 –10 servings/day), and low-fat dairy products (2–3 servings/day); avoiding excessive alcohol consumption (no more than 2 servings/day in men and no more than 1 serving/day in women); and increasing physical activity levels. These nonpharmacological strategies may also positively affect glycemia and lipid control (20). The DASH (Dietary Approaches to Stop Hypertension) diet, which is high in vegetables and fruit, low-fat dairy products, and low in saturated and total fat; has been shown in large, randomized, controlled trials to significantly reduce blood pressure (43).

 

Magnesium

 

Studies in support of magnesium supplementation to improve glycemic control are unclear and complicated by differences in study designs as well as baseline characteristics. There is some evidence from observational data that higher dietary intake of magnesium may help prevent type 2 diabetes in both middle aged men and women at higher risk for developing the disease (44).  Additional long-term studies are needed to determine the best way to assess magnesium status and how magnesium deficiency impacts diabetes management, however dietary sources of magnesium include nuts, whole grains, and green leafy vegetables can be encouraged as part of a healthy dietary pattern.

 

Chromium

 

Several studies have demonstrated a potential role for chromium supplementation in the management of insulin resistance and type 2 diabetes. According to the ADA position statement, the findings with more significant effects were mainly found in poorer quality studies, limiting transferability of the results. Routine supplementation of chromium is therefore not recommended for treating diabetes or obesity (45).

 

HERBAL SUPPLEMENTS

 

There has been interest in the past several years on the effect of cinnamon, curcumin, and other herbs and spices in individuals with diabetes. The most recent ADA Lifestyle Management recommendations conclude that after a review of the evidence, there is not enough clear data to substantiate recommending the use of herbs or spices as treatment for T2D (1). The ADA also states that the use of any herbal supplements, which are not regulated and vary in content, may provide more risk than benefit, in that herbs may interact with other medications that are taken to control diabetes (20).

 

PROBIOTICS

 

Probiotics (from pro and biota, meaning "for life"), are certain kinds of “good” bacteria found in fermented foods, such as yogurt, kefir, and kimchi and are available as supplements. They are naturally found in the gut and may be depleted due to poor diet, use of antibiotics, smoking, etc. Probiotics have been studied extensively to improve gut flora for use in treatment and possibly prevention of various disorders, including irritable bowel syndrome, diarrhea, constipation, and genitourinary infections, to name a few. Different strains and amounts may work better for some conditions over others, but the FDA does not oversee the supplements, so content and effectiveness are not regulated. They are generally considered safe, as they are found naturally in the digestive tract.

 

Some research has been done in people with gestational and type 2 diabetes using probiotic supplements and foods to determine if chronic inflammatory and glycemic markers can be improved (46). The premise is that the microbiota maybe connected to glucose metabolism by altering insulin sensitivity and inflammation. At present there is insufficient evidence to make recommendations for people with diabetes to take a probiotic for glycemic control.

 

ALCOHOL

 

Updated guidelines recommend there is no safe level of alcohol consumption (10). Adults with diabetes who chose to drink alcohol should do so in moderation (no more than one drink per day for adult women and no more than two drinks per day for adult men). Alcohol consumption may place people with diabetes at increased risk for hypoglycemia, especially if taking insulin or insulin secretagogues. Education and awareness regarding the recognition and management of delayed hypoglycemia due to alcohol with or without a meal are warranted. Risks of excessive alcohol intake include hypoglycemia (particularly for those using insulin or insulin secretagogue therapies), weight gain, and hyperglycemia (for those consuming excessive amounts). Hypoglycemia can occur through several mechanisms, including the inability of alcohol to be converted into glucose, the inhibitory effect of alcohol on gluconeogenesis, and its interference in normal counter regulatory hormonal responses to impending hypoglycemia. To decrease the risk of alcohol induced hypoglycemia, it is best to have the alcohol with food. Consuming alcohol in a fasting state may contribute to hypoglycemia in people with type 1 diabetes. Symptoms of hypoglycemia can be similar to drunkenness. When calculating the need for meal related boluses of insulin, one should account for the carbohydrate content of the alcohol if drinking sweet wines, liqueurs, or drinks made with regular juice or soda.

 

PUTTING IT ALL TOGETHER- FOR TYPE 1 DIABETES AND THOSE ON INSULIN

 

People taking insulin should be counseled on the importance of balancing food and beverage intake with timing and dosing of insulin. This is especially important for individuals with varied or hectic schedules such as shift workers, people that travel frequently, or anyone who has a schedule in which timing of meals and access to food is irregular (20).Numerous materials and resources are available that can be provided to people with diabetes to help them consider portion control, consistency in food intake and medication dosing, as well as planning to allow some flexibility in their daily self-care regimen (47). Ongoing support from a referral to medical nutrition therapy conducted by a registered dietitian (RD) or registered dietitian nutritionist (RDN), or a referral to a diabetes self- management education (DSMES) program that includes dietary advice is highly effective. The health care provider should provide individualized guidelines for a target blood glucose range, considering safety and health. For motivated people, teaching an insulin to CHO ratio and blood glucose correction factor may assist them with achieving blood glucose targets and achieving better glycemic control (1).

 

CARBOHYDRATE COUNTING

 

Carbohydrate counting is a tool that can be taught to the motivated, so that they can more easily estimate the amount (grams) of CHO in a particular food and adapt their insulin therapy accordingly (48). Furthermore, setting a target CHO intake for each meal allows those with diabetes to better match their CHO intake to the appropriate mealtime insulin dose. Potential advantages of CHO counting include improved glucose control, flexibility in food choices, a better understanding of how much insulin to take, and simplification of meal planning (49).

 

Carbohydrate (CHO) intake affects acute blood glucose levels. Monitoring carbohydrate, whether by carbohydrate counting, using the exchange method, or experienced- based estimation, remain an important strategy used in timing of medication administration and improving glycemic control (20). CHO counting methodology is based on the concept that each serving of CHO equals approximately 15 grams of CHO. Generally, blood glucose response to digestible carbohydrate is similar, however carbohydrate sources naturally high in fiber including whole grains, legumes, vegetables, and whole fruits should be encouraged over highly processed foods, fruit juices, and sweetened beverages. Insulin dosing also needs to be adjusted based on the protein and fat content of the mail as well, as high levels of either can slow down digestion and glucose uptake into circulation. On average woman require about 3-4 servings (45-60 grams), while men may need 4-5 servings (60-75 grams) of CHO at each meal (47). This number could vary depending on individual energy needs (i.e., pregnant/nursing, ill, etc.), medication, and level of physical activity.

 

A good online resource for basic carbohydrate counting can be found on the UCSF website:

https://dtc.ucsf.edu/living-with-diabetes/diet-and- nutrition/understanding-carbohydrates/counting- carbohydrates/

 

SPECIAL CONSIDERATIONS FOR THOSE WITH INTENSIVE INSULIN REGIMENS

 

The following guidelines are the starting point for the nutritional component of intensified insulin management regimens for those not on closed loop systems: (1, 50)

 

  • The initial diabetes meal plan should be based on the individual’s normal intake with respect to calories, food choices, and times of meals eaten.
  • Choose an insulin regimen that is compatible with their normal pattern of meals, sleep, and physical
  • Synchronize insulin with meal times based on the action time of the insulin(s) used.
  • Assess blood glucose levels prior to meals and snacks and at bedtime and adjust the insulin doses as needed based on intake.
  • Monitor A1C, weight, lipids, blood pressure, and other clinical parameters, modifying the initial meal plan as necessary to meet goals.
  • It is also important to educate those with diabetes on adjustment of prandial insulin considering premeal glucose levels, carbohydrate intake, and anticipated physical activity.
  • For those with diabetes who are overweight and on insulin, counseling on nutrition, weight management, and monitoring blood glucose continues to be important components of treatment. Medical nutrition therapy is recommended with continued emphasis on making lifestyle changes to achieve a weight loss of 5% or more to reduce the risk of chronic complications associated with diabetes, CVD, and other risk factors that contribute to early mortality.

 

CHILDREN AND ADOLESCENTS

 

While medical nutrition therapy provided by registered dietitians resulted in better glycemic control in children with newly diagnosed type 1 diabetes, a survey of 45 pediatric clinics revealed that only 25 clinics had an experienced pediatric/adolescent dietitian available for children with diabetes (51). Registered Dietitian Nutritionists who are trained and experienced with children and adolescent diabetes management should be involved in the multidisciplinary care team (52). The goals of nutrition therapy for children and adolescents with diabetes include the following: (1, 52)

 

  • Provide individualized nutrition therapy with guidance on appropriate energy and nutrient intake to ensure optimal growth and development.
  • Assess and consider changes in food preferences over time and incorporate changes into
  • Promote healthy lifestyle habits while considering and preserving social, cultural, and physiological well-
  • Achieve and maintain the best possible glycemic
  • Achieve and maintain appropriate body weight and promote regular physical activity.

 

Dietary Advice Should Start Gradually

 

    • Emphasis should initially be on establishing supportive rapport with the child and family with simple instructions. More detailed guidelines should be administered later by the entire team, with focus on consistency in message, and should include dietary guidelines to avoid hypoglycemia. Instruction on carbohydrate counting should be provided as soon as possible after diagnosis (52).
    • Nutritional advice needs to be given to all caregivers; babysitters, and extended family who care for the child.
    • Nutrition guidelines should be based on dietary history of the family and child’s meal pattern and habits prior to the diagnosis of diabetes and focus on nutritional recommendations for reducing risk of associated complications and cardiovascular risk that are applicable to the entire family.
    • Physical activity schedules need to be assessed, along with 24-hour recall, and 3-day food diary to determine energy Growth patterns and weight gain need to be assessed every 3-6 months and recommended dietary advice adjusted accordingly (51).

     

    Dietary recommendations can be illustrated by use of the Plate method. There are numerous resources for visuals and educational materials using the plate method and some are specific to diabetes. Half the plate should consist of vegetables and fruit, while the other half is divided between whole grains and lean sources of protein. The dairy is represented by a glass of nonfat or 1% milk or other nonfat or low-fat dairy source. The general guidelines for macronutrients are similar to that of the adult population with diabetes (1, 10).

 

Figure 1. Choosemyplate.gov. Video and print materials can be found on the website.

 

PREVENTION OF HYPOGLYCEMIA

 

Hypoglycemia usually occurs when taking insulin, or when taking a sulfonylurea. To help prevent hypoglycemia, the following guidelines should be discussed:

 

  • Don't skip or delay meals or snacks. If taking insulin or sulfonylurea, be consistent about the amount eaten and the timing of meals and snacks.
  • Monitor blood glucose closely.
  • Measure medication carefully, and take it on time. Take medication as recommended by the physician coordinating diabetes care.
  • Adjust medication or eat additional snacks if physical activity The adjustment depends on the blood glucose test results and on the type and length of the activity.
  • Eat a meal or snack if choosing a drink with alcohol. Drinking alcohol on an empty stomach can contribute to hypoglycemia.
  • Record low glucose reactions. This can help the health care team identify patterns contributing to hypoglycemia and find ways to prevent them.
  • Carry some form of diabetes identification so that in an emergency others will know you have diabetes. Use a medical identification necklace or bracelet and wallet card.

 

SICK DAY MANAGEMENT

 

Eating and drinking can be a challenge when sick. The main rules for sick day management are:

 

  • Continue to take diabetes medication (insulin or oral agent).
  • Self-monitor blood
  • Test urine
  • Eat the usual amount of carbohydrate, divided into smaller meals and snacks if necessary.
  • Drink non-caloric, caffeine free fluids frequently.
  • Call the diabetes care team.

 

See more at: https://diabetes.org/getting-sick-with-diabetes/sick-days

 

PHYSICAL ACTIVITY

 

Regular physical activity has many health benefits. For individuals with diabetes, these benefits outweigh potential risks. Physical activity can improve glycemic control (1). People with diabetes should be encouraged to undertake regular physical activity to improve cardiovascular and overall fitness, weight control, and for improved psychological well-being and quality of life (20). Physical activity can be considered in terms of duration, intensity, modality, and regularity. The largest potential risks due to physical activity for people with diabetes relates most to the intensity of activity. Low intensity physical activity is safer than high intensity physical activity, and highly beneficial for those with diabetes. People with diabetes are encouraged to undertake at least 30 minutes physical activity each day, with modalities such as walking recommended. There is some evidence that walking is more beneficial to glycemia when undertaken within the two hours after meals, (53) as the skeletal muscles take glucose out of circulation to use as fuel. High intensity physical activity should be discussed first with the diabetes care team due to the potential risk of hypoglycemia and cardiovascular strain. To summarize, there are several factors that can affect the blood glucose response to physical activity: (54)

 

  • Individual response to physical activity
  • Duration, intensity, modality, and regularity of physical activity.
  • Timing and type of the previous
  • Timing and type of the insulin injection or other diabetes
  • Pre-physical activity blood glucose
  • Person’s fitness

 

In individuals taking insulin, blood glucose monitoring is necessary to adjust insulin dosing and carbohydrate intake to reduce hypoglycemia due to physical activity. To reduce the risk of hypoglycemia, when higher intensity physical activity is planned, it may be preferable to adjust the dose of insulin before the activity begins. On the other hand, if the physical activity is unplanned, blood glucose should be checked and a carbohydrate snack can be eaten as needed before the activity begins. If the blood glucose is less than 100mg/dL, a 15- to 30-g carbohydrate snack should be consumed, and glucose should be rechecked in 30 to 60 minutes. If glucose levels are less than 70 mg/dL, physical activity should be postponed. Depending on the blood glucose level at the start of physical activity, as well as duration and intensity of the activity, a snack may need to be consumed before, during and after the physical activity. Moderate intensity physical activity can increase glucose uptake significantly, which may require an additional 15 grams of carbohydrate for every 30-60 minutes of exercise above the normal routine (54).

 

Physical activity can increase the rate of absorption of insulin into limbs, especially when it is started immediately after the insulin injection. Inject insulin into a less-active area, such as the abdomen, to minimize the effect of physical activity on insulin absorption. Guidelines for glucose management with exercise exist for those with type 1 diabetes (55). The response to physical activity varies greatly in every individual, so adjustment in medication and food should be based on individual responses. Blood glucose monitoring is very important in understanding response patterns and tailoring a physical activity program (56).

 

TIMING OF INSULIN AND MEALS

 

The greatest risk for hypoglycemia results when the peak insulin action does not coincide with the peak postprandial glucose. For example, the longer duration of action of regular insulin may lead to increased risk of late postprandial hypoglycemia, compared with rapid-acting insulin analogs, which peak closer to meal consumption. In addition, when the pre-meal insulin dose is too large for a particular meal relative to its CHO content, hypoglycemia can result. Such a mismatch may occur due to errors in estimating CHO or food intake. Insulin calculations can be based on exchanges, carbohydrate counting, or predefined, set menus. If meals and the insulin regimen remain constant, then no problems will usually result. However, any changes in insulin or food intake require adjustment of one or the other, or both. Whatever regimen is employed, it must be individualized. Those taking rapid-acting insulin may choose to give their insulin dose after the meal, if unsure of amount of food to be consumed. This approach can be especially helpful in children or in nausea related to pregnancy or illness. If a smaller than normal meal is eaten, guidelines are available for reducing the insulin dose, or carbohydrate replacement in the form of fruit or fruit juice can be given, depending on the particular insulin regimen (57).

 

HYPOGLYCEMIA TREATMENT GUIDELINES

 

Hypoglycemia is defined as a low blood glucose level ≤70 mg/dL. Symptoms include anxiety, irritability, light- headedness and shakiness. Advanced symptoms include headache, blurred vision, lack of coordination, confusion, anger, and numbness in the mouth. Hypoglycemia must be treated immediately with glucose. Follow the 15/15 rule: take15 grams of simple carbohydrate which should increase blood glucose by 30-45 mg/dL within 15 minutes. When blood glucose dips below 70 mg/dL and oral carbohydrate can be administered, have one of the following "quick fix" foods right away to raise the glucose:

 

  • Glucose tablets (see instructions).
  • Gel tube (see instructions).
  • 4 ounces (1/2 cup) of juice or regular soda (not diet).
  • 1 tablespoon of sugar, honey, or corn
  • Hard candies, jellybeans, or gumdrops, see food label for how many to consume.

 

High-fat foods will delay peak of glucose levels from carbohydrate intake and should be avoided (e.g., treatment of hypoglycemia with chocolate bars). After 15 minutes, blood glucose should be checked again to make sure that it is increasing. If it is still too low, another serving is advised. Repeat these steps until blood glucose is at least 70 mg/dL. Then, a snack should be consumed if it will be an hour or more before the next meal.

 

Those who take insulin or a sulfonylurea should be advised to always carry one of the quick-fix foods with them, when driving, and also have available nearby when sleeping. Wearing a medical ID bracelet or necklace is also a good idea, as is having a glucagon emergency kit or nasal spray on hand and knowing how to administer, as well as training close contacts to administer as well.

 

WEIGHT LOSS FOR THOSE WHO WISH TO LOSE WEIGHT

 

While the general principles discussed so far apply to all people with diabetes, those with type 2 diabetes who are overweight or obese (BMI >25 kg/m2 or >23 kg/m2 for Asians) and wish to lose weight can require greater support to do so. Consistent evidence has indicated that intentional weight loss reduces blood glucose in people with type 2 diabetes, and improves most other major cardiometabolic risk factors (58, 59). Clinical guidelines state that weight loss through nutrition and physical activity are fundamental to type 2 diabetes management (60, 61). However, with so many weight loss “diets” available, confusion abounds and reinforces the absolute importance that health professionals provide consistent, evidence-based advice. It is also important to have realistic expectations about the speed at which weight is lost. Obesity does not occur overnight, and its treatment requires long term adjustments to energy intake and expenditure.

 

Many randomized, controlled trials and meta-analyses of trials have been undertaken and to ascertain which macronutrient combination leads to greater weight loss. A two-year head- to-head trial comparing four weight loss diets with differing macronutrient content concluded that all four reduced energy diets, regardless of macronutrient content, led to comparable modest weight loss with weight regain over time (62). This finding was reinforced by a recent comprehensive Cochrane systematic review of RCTs of adults with overweight or obesity with or without type 2 diabetes (11). This review concluded that there is probably little to no difference in weight reduction and changes in cardiovascular risk factors up to two years' follow-up, when overweight and obese participants without and with T2DM are randomized to either low-carbohydrate or balanced-carbohydrate weight-reducing diets. The understanding that focusing on reducing energy intake overall, rather than through a specific macronutrient, frees up weight loss advice so that it can be tailored to the individual’s personal, cultural, and social norms. In this context, understanding reasons for eating, portion size, the energy density of different foods, and factors that promote satiety such as high fiber intakes become essential for achieving and maintaining weight loss.

 

The most important variable in selecting a weight loss plan is the ability of the individual to follow it over the long term. Developing an individualized weight loss program together, preferably with a registered dietitian nutritionist familiar with diabetes management, along with regular follow-ups, will help promote and maintain weight loss. Initial physical activity recommendations alongside dietary changes should be moderate, gradually increasing the duration and frequency to at least 30 minutes a day of activities such as walking.

 

Current evidence indicates that ‘low’, and ‘very low’ energy diets using total or partial diet replacement formula diet products are highly effective for weight loss and reduction of other cardiometabolic risk factors when compared with food-based weight-loss diets (63-65). Furthermore, low-energy nutritionally complete formula diets with a ‘total diet replacement’ induction phase are the most effective dietary approach for achieving type 2 diabetes remission (65-67). Comparing ‘low’ with ‘very-low’ energy diets, many people find very-low-energy diets (420–550 kcal/day) difficult to sustain, and they do not generate greater weight loss than formula diets providing ~810 kcal/day (63, 65).

 

While fast weight loss is a highly desirable outcome, the ultimate health benefits from weight management are likely to depend on long-term weight loss maintenance (10). Long-term low-intensity structured programs, including support for changing food choice, eating pattern and physical activity, and psychological support for behavior change, can help to sustain new behaviors, relationships with foods and adherence to dietary advice, and thus improve weight-loss maintenance (68, 69). Given that dietary adherence can be socially and psychologically testing, skills and empathy from the health professional is needed, providing consistent, long-term, evidence-based support (70). Discussions with patients around weight loss should be entered into with their permission (71) and are important, given the prevalence of obesity (72) and its connection to diabetes incidence (18).

 

MEAL PLANNING APPROACHES

 

There is no one “diet” for diabetes. There are, however, many meal planning guidelines available for the people with diabetes. Listed in the information below are some of the meal planning approaches available.

 

Choose My Plate

 

Choose My Plate contains general, simple guidelines for healthy eating using a small plate to visually illustrate foods and portion control. Print materials and videos from the USDA are available at www.choosemyplate.gov. and The Joslin Diabetes Center https://www.joslin.org/info/diabetes-and-nutrition.html

 

Diabetes Place Mat

Figure 2. Nutrition Place Mat for Diabetes. A sturdy, heavily laminated, 11" by 17" place mat that can be easily used over and over to apply the meal plan.

 

One side of the Diabetes Place Mat lists food choices and individual portion sizes for each food category of the meal plan. This list replaces easily misplaced or damaged paper lists. When planning the meal, a wipe-off marker is used to write down the number of servings for each food category, as indicated on the plan. Then circle or tally the food choices in each category to track progress toward the plan’s targets. Carbohydrate categories - starch and bread, fruit, milk and other carbohydrates - which affect blood glucose and which can be exchanged for each other, are color coded in yellow for easy identification and proper selection. Other food categories - vegetables, meat, fat and free foods - are individually color-coded.

 

The other side of the Diabetes Place Mat illustrates the "Plate Method" of managing a diet for proper nutrition and control of blood glucose and weight. It shows the proportions of each food category that are appropriate for a healthy, balanced diet. The food groups shown on the top half of the Plate Method side are carbohydrates, which affect blood glucose the most - fruit, milk, and starch & bread. These are colored in yellow to distinguish them from the other food groups that don't significantly affect blood glucose (meat, vegetables, fat and free foods). The food categories are shown in proportion to how much of each might be eaten in a healthy, balanced diet. The plate method is a great plan for those who have poor math or reading skills or are non- English speaking.

 

Mediterranean-Style Eating

 

The Mediterranean-style eating pattern derived from the Mediterranean region of the world has been observed to improve glycemic control and cardiovascular disease risk factors. The Mediterranean eating pattern includes:

 

  • Vegetables, fruits, nuts, seeds, legumes, potatoes, whole grains, breads, herbs, spices, fish, seafood and extra virgin olive oil. Emphasis is placed on use of minimally processed foods, seasonal fresh and locally grown foods.
  • Olive oil is the primary fat, replacing other fats and oils (including butter and margarine).
  • Fresh fruit as daily dessert; sweets only
  • Low-to-moderate amounts of cheese and
  • Red meat limited to only 12 oz to 16 oz per month.

 

DASH Eating Plan

 

Dietary Approaches to Stop Hypertension (DASH) is a flexible and balanced eating plan that is based on research studies sponsored by the National Heart, Lung, and Blood Institute (NHLBI). The DASH diet emphasizes vegetables, fruit, fat-free or low-fat dairy, whole grains, nuts and legumes, and limit the intake of total and saturated fat, cholesterol, red and processed meats, sweets and added sugars, including sugar-sweetened beverages. Results from RCTs indicate reductions in glycemia, blood pressure, body weight, and -cholesterol concentrations (73). In prospective cohort studies the DASH diet is associated with reductions in the risk of CVD, CHD and stroke (73). DASH is considerably lower in sodium than the typical American diet.

 

Intermittent Fasting

 

The popularity of intermittent fasting has increased recently as a new way to lose weight and possibly lead to better control of Type 2 diabetes. There are many suggested types of intermittent fasts; some involve eating only on specific days, or not eating for a specified number of hours, alternated by day or hours in which food consumption is allowed. Others greatly restrict energy intake on some days but allow a more normalized diet on other days. There is no one specific intermittent fasting diet that has been proven to be beneficial. Since energy intake is restricted for certain periods of time, an individual with diabetes may lose weight over time if they maintain an overall energy deficit in relation to energy expenditure as is seen with any successful weight loss method.

 

For people with diabetes who are interested in intermittent fasting, current ADA guidance considers time-restricted eating or shortening the eating window adaptable to any eating pattern, and largely safe for adults with type 1 or type 2 diabetes (1, 22). However, anti-hyperglycemic medication use must be considered (74). For those on insulin or taking other anti-hyperglycemia medications, intermittent fasting may lead to hypoglycemic events that may become severe when medications are not adjusted down (75). Careful monitoring of blood glucose is required, and medication adjustment may be necessary. Overall, the simplicity of intermittent fasting and time-restricted eating may make these useful strategies for people with diabetes who are looking for practical eating management tools (1).

 

Gluten Free

 

Gluten is a protein commonly found in wheat, barley, rye, and other grains. A gluten free diet is essential to treat people with celiac disease. Celiac disease is an inflammatory condition in persons who are intolerant to gluten and suffer inflammatory and gastrointestinal side effects when gluten is consumed, leading to damage of the small intestine. It is noted that approximately 10% of people with type 1 diabetes also have celiac disease, which is significantly higher than the general population (1-2%). There seems to be no connection with Celiac disease and type 2 diabetes (76). There is no evidence of health benefits when avoiding gluten for those without celiac disease.

 

The gluten free diet has recently grown in popularity in persons who identify as gluten sensitive, but don’t have celiac disease. According to the ADA, people with T1D can follow a gluten free diet should they wish to, but it may provide additional challenges. Common CHO containing foods that do not contain gluten are: white and sweet potatoes, brown and wild rice, corn, buckwheat, soy, quinoa, sorghum, and legumes. These foods can be used in place of gluten containing grains.

 

MAYO Clinic Diet

 

Developed by the Mayo Clinic, a two-phase approach to lose and maintain body weight using the Mayo Clinic food pyramid. For more information see: https://diet.mayoclinic.org/diet/how-it-works.

 

Jenny Craig®

 

The plan emphasizes restricting energy, fat, and portions. Jenny's prepackaged meals and recipes do all three, plus emphasize healthy eating, an active lifestyle, and behavior modification. Personal consultants guide members through their journeys from day one. You'll gain support and motivation, and learn how much you should be eating, what a balanced meal looks like and how to use that knowledge once you graduate from the program. Jenny Craig offers two programs: its standard program and Jenny Craig for Type 2, which is designed for people with Type 2 diabetes by including a lower-carb menu, reinforcement of self-monitoring of blood glucose levels, consistent meals and snacks, and other self-management strategies for weight loss and support for diabetes control. Because you buy foods, this program can be more expensive, but convenient for some. For more information see: https://www.jennycraig.com/.

 

Vegan Diet

 

Veganism excludes all animal products from the diet – including dairy and eggs. Fruits, vegetables, leafy greens, wholegrains, nuts, seeds and legumes are the staples. It is restrictive, but beneficial for the cardiovascular system.

 

Weight Watchers®

 

The Weight Watchers assigns every food and beverage a point value, based on nutritional content and provides users with a maximum number of points they can consume per day. A backbone of the plan is multi-model access (via in-person meetings, online chat or phone) to support from people who lost weight using Weight Watchers, kept it off and have been trained in behavioral weight management techniques. For more information see: https://health.usnews.com/best-diet/weight-watchers-diet

 

Individualized Menus Provided by a RD/RDN

 

Many people with diabetes might like to have examples to follow when setting up meal plans. The menu describes in writing what foods and what quantities should be consumed over a period of days. A dietitian creates an individualized menu based on the specific nutritional counseling plan and incorporates the client’s unique preferences, schedule, etc. The client then has written examples to follow, and over time may learn how to independently create their own menus and substitutions to fit their individual lifestyle.

 

Month of Meals

 

These menus were created by committees of the Council on Nutritional Science and Metabolism of the American Diabetes Association, and staff of the American Diabetes Association National Service Center in response to frequent requests for menus from people with diabetes and their families. The menus are designed to follow the exchange groups and provide 45-50% of energy from CHO, 20% protein, and about 30% fat. The menus provide 1200 or 1800 calories, and instructions are provided on how to adjust caloric levels upward or downward. Each menu provides 28 days of breakfast, lunch, dinner and snacks with a different focus to help make planning meals easier.

 

Exchange List Approach

 

The Exchange Lists for Meal Planning were developed by the American Diabetes Association and the Academy of Nutrition and Dietetics, and have been in existence since 1950. The latest version of Choose Your Foods: Food Lists for Diabetes was released in 2019. The concept for this list is that foods are grouped according to similar nutritional value, and can be exchanged or substituted in the portion size listed within the same group. The exchange lists include:

 

  • Carbohydrate group – includes starches, fruit, milk and
  • Meat and Meat Substitutes group – four meat categories based on the amount of fat they
  • Fat group – contains three categories of fats based on the major source of fat contained: saturated, polyunsaturated or monounsaturated.

 

The exchange lists also give information on fiber and sodium content. They can be utilized for people with type 1 or 2diabetes. The emphasis for type 1 is on consistency of timing and amount of food eaten, while for type 2, the focus is on controlling the caloric values of food consumed. Use of the exchange list may be helpful for some people while others may benefit by learning from other carbohydrate counting resources available online and through numerous publications and resources.

 

Calorie Counting

 

These are meal planning methods that can be useful for people with type 2 diabetes who want to lose weight. Knowledge regarding the number of total calories in a given food (including pre-prepared and fast foods) and becoming adept at label reading, can help promote weight loss when incorporated into other lifestyle changes. One of the first studies designed to determine empirically if people can learn a calorie counting system and if estimated food intake improves with training demonstrated that use of the Health Management Resources Calorie System tool (HMRe, Boston, MA, USA) helped to teach people how to estimate food intake more accurately (77).

 

RESOURCES FOR DIABETES NUTRITION EDUCATION

 

Table 3. DIABETES NUTRITION EDUCATION RESOURCES

Choose My Plate

www.choosemyplate.gov

Eat Out, Eat Well

 

Your go-to resource for assembling healthy meals in just about any type of restaurant, from fast food to upscale dining and ethnic cuisines. Order from: The American Diabetes Assn., www.shopdiabetes.org, 1-800-232-6455

American Diabetes Associate: Standards of Care

Facilitating positive health behaviors and well-being to improve health outcomes: standards of Care in Diabetes. 2024 https://doi.org/10.2337/dc24-SINT

DNSG European Dietary Guidelines

The Diabetes Nutrition Study Group of the European Association for the Study of Diabetes. Diabetologia. 2023 https://doi.org/10.1007/s00125-023-05894-8

What Can I Eat? The Diabetes Guide to Healthy Food Choices2nd Edition

A 28-page guide for planning meals and making the best food choices. Includes carb counting, glycemic index, plate method, eating out, meals/snack ideas, best food choices and more. Order from: The American Diabetes Assn., Inc. www.shopdiabetes.org, 1-800-232-6455

Eating Healthy with Diabetes, 5th Edition

Picture cues for portion sizes and color codes for food types teach how to put together a healthy diet plan to manage diabetes Order from: The Academy of Nutrition and Dietetics. www.eatright.org or the American Diabetes Assn., Inc. www.shopdiabetes.org.

Diabetes Meal Planning Made Easy & Healthy Portions Meal Measure

Meet your health and nutrition goals with healthy diabetes meal plans, shopping strategies and our handy portion control guide. Order from: The American Diabetes Association, www.shopdiabetes.org, 800-232-6455

Diabetes Place Mat Kit for People with Diabetes

Order from: NCES Health & Nutrition Information Catalog- Available inSpanish https://www.ncescatalog.com/NCES- MyPlacemat-for-Diabetes_p_1103.html OR

School Health Corporation https://www.schoolhealth.com/nutrition-place-mat-for-diabetes

The Complete Month of Meals Collection, 2017

Available from: Amazon.com or American Diabetes Association, 1-800-232-6455; www.shopdiabetes.org

Choose Your Foods: Food Lists for Diabetes

Order from: Academy of Nutrition and Dietetics OR American Diabetes Associations; www.eatright.org OR http://shopdiabetes.org or Amazon.com Available in Spanish

Diabetes Food Hub

www.diabetesfoodhub.org. A website available on the American DiabetesAssociation site that has meal planning, grocery lists, recipes, menus and healthy substitutions. Section in Spanish available.

The Complete Guide to Carb Counting

American Diabetes Association 4th edition. Has all the expert information you need to practice carb counting, whether you’re learning the basics or trying to master more advanced techniques. Order from American Diabetes Association, http://shopdiabetes.org or Amazon.com

Diabetes and CarbCounting for Dummies1st Edition

By Sherri Shafer, RD, CDE. Provides essential information on how to strike a balance between carb intake, exercise, and diabetes medications while making healthy food choices. Available at Amazon.com

 

The resources listed above are a sampling of the many available, primarily from the American Academy of Nutrition and Dietetics and the American Diabetes Association. There are several other organizations and websites which have educational materials available:

 

REFERENCES

 

  1. American Diabetes Association. 5. Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes: Standards of Care in Diabetes—2024. Diabetes Care. 2024;47(Supplement 1):S77-S110.
  2. Fang M, Wang D, Coresh J, Selvin E. Trends in Diabetes Treatment and Control in U.S Adults, 1999-2018. New England Journal of Medicine. 2021;384(23):2219-2228.
  3. Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, et al. American Association of Clinical Endocrinology Clinical Practice Guideline: Developing a Diabetes Mellitus Comprehensive Care Plan: 2022 Update. Endocrine Practice. 2022;28(10):923-1049.
  4. The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. New England journal of medicine. 1993;329(14):977-86.
  5. World Health Organization. Guideline: sugars intake for adults and children: World Health Organization; 2015.
  6. Reynolds A, Mann J, Cummings J, Winter N, Mete E, Te Morenga L. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet. 2019;393(10170):434-45.
  7. World Health Organization. Carbohydrate intake for adults and children: WHO guideline. Carbohydrate intake for adults and children: WHO guideline2023.
  8. Reynolds AN, Akerman AP, Mann J. Dietary fibre and whole grains in diabetes management: Systematic review and meta-analyses. PLoS medicine. 2020;17(3):e1003053.
  9. Reynolds AN, Akerman A, Kumar S, Diep Pham HT, Coffey S, Mann J. Dietary fibre in hypertension and cardiovascular disease management: systematic review and meta-analyses. BMC Medicine. 2022;20(1):139.
  10. The Diabetes Nutrition Study Group of the European Association for the Study of Diabetes (DNSG). Evidence-based European recommendations for the dietary management of diabetes. Diabetologia. 2023;66(6):965-85.
  11. Naude CE, Brand A, Schoonees A, Nguyen KA, Chaplin M, Volmink J. Low‐carbohydrate versus balanced‐carbohydrate diets for reducing weight and cardiovascular risk. Cochrane Database of Systematic Reviews. 2022(1).
  12. Reynolds AN, Diep Pham HT, Montez J, Mann J. Dietary fibre intake in childhood or adolescence and subsequent health outcomes: A systematic review of prospective observational studies. Diabetes, Obesity and Metabolism. 2020;22(12):2460-7.
  13. Kelly RK, Tong TYN, Watling CZ, Reynolds A, Piernas C, Schmidt JA, et al. Associations between types and sources of dietary carbohydrates and cardiovascular disease risk: a prospective cohort study of UK Biobank participants. BMC Medicine. 2023;21(1):34.
  14. Åberg S, Mann J, Neumann S, Ross AB, Reynolds AN. Whole-Grain Processing and Glycemic Control in Type 2 Diabetes: A Randomized Crossover Trial. Diabetes Care. 2020;43(8):1717-23.
  15. Gomes F, Reynolds A, editors. Effects of Fruits and Vegetables Intakes on Direct and Indirect Health Outcomes, Background Paper for the FAO/WHO International Workshop on fruits and vegetables; 2020.
  16. Mann J, Truswell S, Hodson L. Essentials of Human Nutrition 6e. Oxford, UK: Oxford University Press; 2023 15 September 2023.
  17. Schwingshackl L, Schwedhelm C, Hoffmann G, Boeing H. Potatoes and risk of chronic disease: a systematic review and dose–response meta-analysis. European journal of nutrition. 2019;58:2243-51.
  18. Teufel F, Seiglie JA, Geldsetzer P, Theilmann M, Marcus ME, Ebert C, et al. Body-mass index and diabetes risk in 57 low-income and middle-income countries: a cross-sectional study of nationally representative, individual-level data in 685 616 adults. The Lancet. 2021;398(10296):238-48.
  19. Cozma AI, Sievenpiper JL, De Souza RJ, Chiavaroli L, Ha V, Wang DD, et al. Effect of fructose on glycemic control in diabetes: a systematic review and meta-analysis of controlled feeding trials. Diabetes care. 2012;35(7):1611-20.
  20. Evert AB, Boucher JL, Cypress M, Dunbar SA, Franz MJ, Mayer-Davis EJ, et al. Nutrition therapy recommendations for the management of adults with diabetes. Diabetes care. 2014;37(Supplement 1):S120-S43.
  21. Fitch C, Keim KS. Position of the Academy of Nutrition and Dietetics: use of nutritive and nonnutritive sweeteners. Journal of the Academy of Nutrition and Dietetics. 2012;112(5):739-58.
  22. Nichol AD, Holle MJ, An R. Glycemic impact of non-nutritive sweeteners: a systematic review and meta-analysis of randomized controlled trials. European journal of clinical nutrition. 2018;72(6):796-804.
  23. World Health Organization. Use of non-sugar sweeteners: WHO guideline. Geneva: World Health Organization; 2023. Licence: CC BY-NC-SA 3.0 IGO.
  24. World Health Organization. Saturated fatty acid and trans-fatty acid intake for adults and children: WHO guideline. Saturated fatty acid and trans-fatty acid intake for adults and children: WHO guideline 2023.
  25. Reynolds AN, Hodson L, De Souza R, Tran Diep Pham H, Vlietstra L, Mann J. Saturated fat and trans-fat intakes and their replacement with other macronutrients: a systematic review and meta-analysis of prospective observational studies. 2022.
  26. Eckel RH, Jakicic JM, Ard JD, Jesus JMd, Miller NH, Hubbard VS, et al. 2013 AHA/ACC Guideline on Lifestyle Management to Reduce Cardiovascular Risk. Circulation. 2014;129(25 suppl 2):S76-S99.
  27. Vannice G, Rasmussen H. Position of the academy of nutrition and dietetics: dietary fatty acids for healthy adults. Journal of the Academy of Nutrition and Dietetics. 2014;114(1):136-53.
  28. Sacks FM, Lichtenstein AH, Wu JH, Appel LJ, Creager MA, Kris-Etherton PM, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1-e23.
  29. Qian F, Korat AA, Malik V, Hu FB. Metabolic effects of monounsaturated fatty acid–enriched diets compared with carbohydrate or polyunsaturated fatty acid–enriched diets in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Diabetes care. 2016;39(8):1448-57.
  30. Schwab U, Reynolds AN, Sallinen T, Rivellese AA, Risérus U. Dietary fat intakes and cardiovascular disease risk in adults with type 2 diabetes: a systematic review and meta-analysis. European Journal of Nutrition. 2021;60(6):3355-63.
  31. Imamura F, Micha R, Wu JH, de Oliveira Otto MC, Otite FO, Abioye AI, et al. Effects of Saturated Fat, Polyunsaturated Fat, Monounsaturated Fat, and Carbohydrate on Glucose-Insulin Homeostasis: A Systematic Review and Meta-analysis of Randomised Controlled Feeding Trials. PLoS Med. 2016;13(7):e1002087.
  32. Van Horn L, McCoin M, Kris-Etherton PM, Burke F, Carson JAS, Champagne CM, et al. The evidence for dietary prevention and treatment of cardiovascular disease. Journal of the American dietetic association. 2008;108(2):287-331.
  33. Mensink RP, Zock PL, Kester AD, Katan MB. Effects of dietary fatty acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr. 2003;77(5):1146-55.
  34. Gupta A, Savopoulos C, Ahuja J, Hatzitolios A. Role of phytosterols in lipid-lowering: current perspectives. QJM: An International Journal of Medicine. 2011;104(4):301-8.
  35. Bard J-M, Paillard F, Lecerf J-M. Effect of phytosterols/stanols on LDL concentration and other surrogate markers of cardiovascular risk. Diabetes & metabolism. 2015;41(1):69-75.
  36. Lok CE, Huber TS, Lee T, Shenoy S, Yevzlin AS, Abreo K, et al. KDOQI clinical practice guideline for vascular access: 2019 update. American Journal of Kidney Diseases. 2020;75(4):S1-S164.
  37. Reynolds AN, Mhurchu CN, Kok Z-Y, Cleghorn C. The neglected potential of red and processed meat replacement with alternative protein sources: Simulation modelling and systematic review. Eclinicalmedicine. 2023;56:101774.
  38. World Health Organization. Red and processed meat in the context of health and the environment: many shades of red and green. Information brief Geneva: World Health Organization. 2023.
  39. Clark MA, Springmann M, Hill J, Tilman D. Multiple health and environmental impacts of foods. Proceedings of the National Academy of Sciences. 2019.
  40. Valdés-Ramos R, Ana Laura G-L, Beatriz Elina M-C, Alejandra Donaji B-A. Vitamins and type 2 diabetes mellitus. Endocrine, Metabolic & Immune Disorders-Drug Targets (Formerly Current Drug Targets-Immune, Endocrine & Metabolic Disorders). 2015;15(1):54-63.
  41. Seida JC, Mitri J, Colmers IN, Majumdar SR, Davidson MB, Edwards AL, et al. Effect of vitamin D3 supplementation on improving glucose homeostasis and preventing diabetes: a systematic review and meta-analysis. The Journal of Clinical Endocrinology & Metabolism. 2014;99(10):3551-60.
  42. World Health Organization. Guideline: Sodium intake for adults and children: World Health Organization; 2012.
  43. Saneei P, Salehi-Abargouei A, Esmaillzadeh A, Azadbakht L. Influence of Dietary Approaches to Stop Hypertension (DASH) diet on blood pressure: a systematic review and meta-analysis on randomized controlled trials. Nutrition, metabolism and cardiovascular diseases. 2014;24(12):1253-61.
  44. Hruby A, Meigs JB, O’Donnell CJ, Jacques PF, McKeown NM. Higher magnesium intake reduces risk of impaired glucose and insulin metabolism and progression from prediabetes to diabetes in middle-aged americans. Diabetes care. 2014;37(2):419-27.
  45. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. 5. Facilitating positive health behaviors and well-being to improve health outcomes: standards of Care in Diabetes 2023. Diabetes Care. 2023;46(Supplement 1):S68-S96.
  46. Shah NJ, Swami OC. Role of probiotics in diabetes: a review of their rationale and efficacy. Diabetes. 2017;5:104-10.
  47. Care D, Education R. Set Start Counting! Carbohydrate Counting–a Tool to Help Manage Your Blood Glucose. Diabetes Care and Education, a dietetic practice group of the Academy of Nutrition and Dietetics. 2016.
  48. Warshaw HS, Kulkarni K. Complete Guide to Carb Counting: How to Take the Mystery Out of Carb Counting and Improve Your Blood Glucose Control: American Diabetes Association; 2011.
  49. Vaz EC, Porfírio GJM, Nunes HRdC, Nunes-Nogueira VdS. Effectiveness and safety of carbohydrate counting in the management of adult patients with type 1 diabetes mellitus: a systematic review and meta-analysis. Archives of endocrinology and metabolism. 2018;62:337-45.
  50. Wolpert HA. Intensive diabetes management: American Diabetes Association; 2016.
  51. Virtanen SM. Medical nutrition therapy of children and adolescents with diabetes. Diabetes in childhood and adolescence: Pediatr Adolesc Med Basel, Basel; 2005. p. 139-49.
  52. Smart CE, Annan F, Higgins LA, Jelleryd E, Lopez M, Acerini CL. ISPAD Clinical Practice Consensus Guidelines 2018: Nutritional management in children and adolescents with diabetes. Pediatric diabetes. 2018;19:136-54.
  53. Reynolds AN, Mann JI, Williams S, Venn BJ. Advice to walk after meals is more effective for lowering postprandial glycaemia in type 2 diabetes mellitus than advice that does not specify timing: a randomised crossover study. Diabetologia. 2016;59(12):2572-8.
  54. Shugart C, Jackson J, Fields KB. Diabetes in sports. Sports Health. 2010;2(1):29-38.
  55. Moser O, Riddell MC, Eckstein ML, Adolfsson P, Rabasa-Lhoret R, van den Boom L, et al. Glucose management for exercise using continuous glucose monitoring (CGM) and intermittently scanned CGM (isCGM) systems in type 1 diabetes: position statement of the European Association for the Study of Diabetes (EASD) and of the International Society for Pediatric and Adolescent Diabetes (ISPAD) endorsed by JDRF and supported by the American Diabetes Association (ADA). Pediatr Diabetes. 2020 Dec;21(8):1375-1393.
  56. Powers MA. Handbook of diabetes medical nutrition therapy: Jones & Bartlett Learning; 1996.
  57. Kaufman FR. Medical management of type 1 diabetes: American Diabetes Association; 2012.
  58. Haase CL, Lopes S, Olsen AH, Satylganova A, Schnecke V, McEwan P. Weight loss and risk reduction of obesity-related outcomes in 0.5 million people: evidence from a UK primary care database. International Journal of Obesity. 2021;45(6):1249-58.
  59. Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni AG, et al. Benefits of Modest Weight Loss in Improving Cardiovascular Risk Factors in Overweight and Obese Individuals With Type 2 Diabetes. Diabetes Care. 2011;34(7):1481-6.
  60. American Diabetes Association. 8. Obesity Management for the Treatment of Type 2 Diabetes: Standards of Medical Care in Diabetes-2021. Diabetes Care. 2021;44(Suppl 1):S100-S10.
  61. Dyson PA, Twenefour D, Breen C, Duncan A, Elvin E, Goff L, et al. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes. Diabet Med. 2018;35(5):541-7.
  62. Sacks FM, Bray GA, Carey VJ, Smith SR, Ryan DH, Anton SD, et al. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. New England Journal of Medicine. 2009;360(9):859-73.
  63. Christensen P, Bliddal H, Riecke BF, Leeds AR, Astrup A, Christensen R. Comparison of a low-energy diet and a very low-energy diet in sedentary obese individuals: a pragmatic randomized controlled trial. Clinical Obesity. 2011;1(1):31-40.
  64. Noronha JC, Nishi SK, Braunstein CR, Khan TA, Blanco Mejia S, Kendall CWC, et al. The Effect of Liquid Meal Replacements on Cardiometabolic Risk Factors in Overweight/Obese Individuals With Type 2 Diabetes: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Diabetes Care. 2019;42(5):767-76.
  65. Churuangsuk C, Hall J, Reynolds A, Griffin SJ, Combet E, Lean ME. Diets for weight management in adults with type 2 diabetes: an umbrella review of published meta-analyses and systematic review of trials of diets for diabetes remission. Diabetologia. 2022;65:14-36.
  66. Thom G, Messow CM, Leslie W, Barnes A, Brosnahan N, McCombie L, et al. Predictors of type 2 diabetes remission in the Diabetes Remission Clinical Trial (DiRECT). Diabetic Medicine. 2021;38(8):e14395.
  67. Lean ME, Leslie WS, Barnes AC, Brosnahan N, Thom G, McCombie L, et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet. 2018;391(10120):541-51.
  68. Ismail K, Winkley K, Rabe-Hesketh S. Systematic review and meta-analysis of randomised controlled trials of psychological interventions to improve glycaemic control in patients with type 2 diabetes. Lancet. 2004;363(9421):1589-97.
  69. American Diabetes Association. 5. Facilitating Behavior Change and Well-being to Improve Health Outcomes: Standards of Medical Care in Diabetes-2021. Diabetes Care. 2021;44(Suppl 1):S53-S72.
  70. Dambha-Miller H, Day A, Kinmonth AL, Griffin SJ. Primary care experience and remission of type 2 diabetes: a population-based prospective cohort study. Fam Pract. 2021;38(2):141-6.
  71. Wharton S, Lau DC, Vallis M, Sharma AM, Biertho L, Campbell-Scherer D, et al. Obesity in adults: a clinical practice guideline. Cmaj. 2020;192(31):E875-E91.
  72. Phelps NH, Singleton RK, Zhou B, Heap RA, Mishra A, Bennett JE, et al. Worldwide trends in underweight and obesity from 1990 to 2022: a pooled analysis of 3663 population-representative studies with 222 million children, adolescents, and adults. The Lancet. 2024;403(10431):1027-50.
  73. Chiavaroli L, Viguiliouk E, Nishi SK, Blanco Mejia S, Rahelić D, Kahleová H, et al. DASH dietary pattern and cardiometabolic outcomes: an umbrella review of systematic reviews and meta-analyses. Nutrients. 2019;11(2):338.
  74. Varady KA, Cienfuegos S, Ezpeleta M, Gabel K. Clinical application of intermittent fasting for weight loss: progress and future directions. Nature Reviews Endocrinology. 2022;18(5):309-21.
  75. Malinowski B, Zalewska K, Węsierska A, Sokołowska MM, Socha M, Liczner G, et al. Intermittent fasting in cardiovascular disorders—an overview. Nutrients. 2019;11(3):673.
  76. Kylökäs A, Kaukinen K, Huhtala H, Collin P, Mäki M, Kurppa K. Type 1 and type 2 diabetes in celiac disease: prevalence and effect on clinical and histological presentation. BMC Gastroenterology. 2016;16(1):76.
  77. Martin CK, Anton SD, York-Crowe E, Heilbronn LK, VanSkiver C, Redman LM, et al. Empirical evaluation of the ability to learn a calorie counting system and estimate portion size and food intake. British Journal of Nutrition. 2007;98(2):439-44.

Normal and Abnormal Puberty

ABSTRACT

 

Puberty is a biological process that represents the development of secondary sexual characteristics and attainment of reproductive capacity, influenced by genetic, metabolic, environmental, ethnic, geographic, and economic factors. Pubertal onset is characterized by the increased kisspeptin and neurokinin B secretion leading to re-emergence of pulsatile gonadotropin releasing hormone signaling from the hypothalamus which stimulates increased pituitary secretion of luteinizing hormone and follicle stimulating hormone, which in turn stimulate gonadal sex hormone production. Precocious puberty refers to secondary sexual development occurring earlier than the lower end of normal age and delayed puberty refers to secondary sexual development occurring later than the upper end of normal age for the onset of puberty. These changes may represent a serious underlying condition or signify a common variation of normal for which treatment may not be necessary. Clinical evaluation should include a detailed history and physical examination, including anthropometric measurements, calculation of linear growth velocity, and evaluation of secondary sexual characteristics. This chapter summarizes the physiology of pubertal development, variations in pubertal development, and recent developments regarding human puberty.

 

INTRODUCTION

 

Puberty is the process through which reproductive competence is achieved (1). Physical characteristics associated with this process include the development of secondary sex characteristics, acceleration in linear growth velocity, and the occurrence of menarche in women and spermatogenesis in men. The sex chromosome karyotype of the embryo, XX or XY, determines the trajectory for differentiation of the gonads and development of the internal and external genital structures. This complex process, beginning in utero, depends on neuroendocrine signaling and gonadal components. Ultimately, integrated communication between the reproductive and metabolic systems is critical for timely pubertal development (2).

 

Pubertal development and neuroendocrine system maturation involve the ontogeny, activity, and interactions of the gonadotropin releasing hormone (GnRH) neurons. The onset of puberty is accompanied by increased kisspeptin and neurokinin B secretion causing the GnRH neurons to secrete GnRH in a pulsatile manner. Increased GnRH secretion stimulates pulsatile pituitary luteinizing hormone (LH) and follicle stimulating hormone (FSH) secretion (3). LH and FSH stimulate gonadal sex steroid secretion which promotes development of secondary sex characteristics and influences hypothalamic-pituitary function via negative feedback inhibition. This chapter summarizes the physiology of pubertal development, variations in pubertal development, and recent developments regarding human puberty.

 

CLINICAL FEATURES OF NORMAL PUBERTAL DEVELOPMENT

 

Children typically demonstrate a predictable sequence of physical changes during pubertal maturation. Within the chronologic age ranges for pubertal development, individual variations regarding age at onset and tempo of pubertal development are expected.

 

In humans, two physiological processes, gonadarche and adrenarche, govern pubertal transition. Gonadarche reflects the reactivation of the hypothalamic GnRH pulse generator which has been quiescent since late infancy. Increasing pulsatile GnRH secretion stimulates pulsatile gonadotropin secretion which, in turn, stimulates the growth and maturation of the gonads and gonadal sex steroid secretion. Increased estrogen secretion promotes breast development, cornification of the vaginal mucosa, and uterine growth in girls. Increased testosterone secretion promotes penile enlargement. The increased HPG axis activity culminates in folliculogenesis, ovulation, and menses in the female and spermatogenesis in the male.

 

In addition to gonadal sex steroid secretion, humans experience adrenarche signifying adrenal pubertal maturation. Adrenarche typically begins prior to the first visible physical manifestation of gonadarche, breast development, or testicular enlargement. Pubarche, the physical manifestation of adrenarche, is characterized by the development of pubic hair, axillary hair, apocrine odor, and acne. Adrenarche indicates increased adrenal cortical zona reticularis activity and is accompanied by increased secretion of dehydroepiandrosterone sulfate (DHEAS), dehydroepiandrosterone (DHEA), androstenedione, and 11-hydroxyandrostenedione (4, 5). These so-called “adrenal androgens” are C19 steroids which do not bind directly to the androgen receptor and can be peripherally converted to more potent androgens. Circulating concentrations of two adrenal 11-oxyandrogens, 11-hydroxyandrostenedione and 11-ketotestosterone increase with adrenarche. Whereas 11-hydroxyandrostenedione has minimal androgenic activity, 11-ketotestosterone is almost as potent as testosterone. During adrenarche, 11-ketotestosterone appears to be the major bioactive adrenal C19 steroid and may be responsible for the physical changes associated with pubarche (6).

 

Gonadarche and adrenarche are dissociated such that the absence of adrenarche does not prevent gonadarche or the attainment of fertility (7). Curiously, the phenomenon of adrenarche appears to be limited to humans and a few species of non-human primates (8, 9). The factors that drive the dynamic changes within a strictly defined zona reticularis within the adrenal cortex, are still poorly defined. How adrenarche and increased adrenal C19 steroids impact brain development during human adolescence is indeterminate (10). Urinary steroid hormone profiling suggest that adrenarche may be a gradual process that likely begins earlier than previously believed (11).

 

STAGING OF PUBERTY

 

Tanner and colleagues followed the pubertal development of children living in an orphanage in the UK. Their five-stage classification system continues to be commonly utilized for clinical assessments (12, 13, 14). For girls, Tanner staging is used to describe breast and pubic hair development (See Figure 1). For boys, Tanner staging is used to describe testicular volume, penile development, and pubic hair development (See Figure 2). Tanner and his colleagues also described that the tempo of puberty varies between individuals.

 

Figure 1. Tanner Staging for pubertal development in girls. In girls, breast development is rated from 1 (preadolescent) to 5 (mature), and stage 2 (appearance of the breast bud) marks the onset of pubertal development. Pubic hair stages are rated from 1 (preadolescent, no pubic hair) to 5 (adult), and stage 2. Although pubic hair and genital or breast development are represented as synchronous in the illustration, they do not necessarily track together and should be scored separately. Figure 1 Adapted with permission from Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2367 and Klein DA, Emerick JE, Sylvester JE, Vogt KS. Disorders of Puberty: An Approach to Diagnosis and Management. Am Fam Physician. 2017 Nov 1;96(9):590-599. PMID: 29094880

Figure 2. Tanner Staging for pubertal development in boys. In boys, genital development is rated from 1 (preadolescent) to 5 (adult); stage 2 marks the onset of pubertal development and is characterized by an enlargement of the scrotum and testis and by a change in the texture and a reddening of the scrotal skin. Pubic hair stages are rated from 1 (preadolescent, no pubic hair) to 5 (adult), and stage 2 marks the onset of pubic hair development. Although pubic hair and genital or breast development are represented as synchronous in the illustration, they do not necessarily track together and should be scored separately. In normal boys, stage 2 pubic hair develops at an average of 12 to 20 months after stage 2 genital development. Figure 2 Adapted with permission from Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2367 and Klein DA, Emerick JE, Sylvester JE, Vogt KS. Disorders of Puberty: An Approach to Diagnosis and Management. Am Fam Physician. 2017 Nov 1;96(9):590-599. PMID: 29094880

 

Girls

 

The typical first clinical sign of puberty in girls is the appearance of breast tissue with elevation of the breast and papilla; this is considered to be Tanner Stage 2 (Figure 1). Initially, breast development (thelarche) may be unilateral. Many girls complain of mild breast tenderness or discomfort during this stage that subsequently resolves. Tanner stage 3 breast development is considered to be additional enlargement of the breast and areola. During Tanner stage 4, the papilla forms a secondary mound above the breast; this stage is often very rapid. Tanner stage 5 represents mature breast development due to recession of the areola to the contour of the breast. Palpation of the breast is obligatory to differentiate breast tissue from lipomastia. In children with obesity without breast development, a palpable depression beneath the nipple in the center of the areola when examined in the supine position gives the impression of a donut and is referred to as the ‘donut’ sign.  Breast ultrasound correlates reasonably well with Tanner staging by palpation and can detect breast development slightly earlier than physical exam (15). In most instances, breast development is evident before pubic hair development. Typically, the pubertal growth spurt in girls occurs concurrently with the onset of breast development with only 4-6 cm of linear growth occurring after menarche, however this may be variable.

 

The appearance of sexual hair including pubic hair (pubarche) signifies the onset of adrenarche. In girls, Tanner stage 2 pubic hair is characterized by sparse, coarse, lightly pigmented hairs along the labia majora. For Tanner stage 3, pubic hair becomes progressively darker, coarser, and spreads over the mons pubis. For Tanner stage 4, pubic hair continues to extend to become an inverse triangle, with spread to the medial aspects of the thighs being considered Tanner stage 5.

 

With the onset of ovarian estrogen secretion, the vaginal mucosa changes from shiny bright red to pale pink appearance due to cornification of the vaginal mucosa. Increased estrogen secretion promotes uterine growth and causes physiologic leukorrhea, a thin, white, non-foul-smelling vaginal discharge that typically begins 6 to 12 months before menarche. Menarche occurs, on average, 2 to 2.5 years after the onset of breast development (See figure 3A). During the first-year post-menarche, menses are usually irregular and anovulatory. These early years are characterized by inconsistent ovulation and varying lengths of follicular and luteal phases. Ultimately, coordinated maturation of the hypothalamic, pituitary, and ovarian components occurs culminating in cyclic monthly ovulation (16). Although full HPG axis maturation generally occurs over several years, by three years post-menarche, most cycles are between 21-35 days.

 

Figure 3A and 3B. Average ages and sequence of pubertal development. Panel A: girls; Panel B: boys.

 

Boys

 

For boys, increased testicular volume is the first physical finding indicating onset of gonadarche (See Figure 2 and Figure 3B). Palpation of the testes and use of a Prader orchidometer is essential for accurate assessment. A Prader orchidometer is a collection of 3-D ellipsoids ranging in volume from 1 to 25 mL (See Figure 4). During gonadarche, testicular volume increases, and the penis increases in length and diameter. Flaccid penile length can be measured using a straight edge on the dorsal surface from the pubic ramus to the tip of the glans while compressing the suprapubic fat pad and applying gentle traction to stretch to penis.

 

Figure 4. Prader Orchidometer.

 

Increased testicular volume represents Sertoli cell proliferation, differentiation, and eventually, the initiation of spermatogenesis. The onset of puberty is defined as a testicular volume > 4 ml and a testicular length > 2.5 cm. The volume of mature human testis is approximately 20-25 ml. Spermatozoa (spermaturia) can be found in early morning urine samples beginning during genital stage 3 (16). Nocturnal sperm emissions may also begin around this time.

 

For boys, Tanner stage 2 pubic hair consists of downy hairs at the base of the penis. During pubic hair stage 3, the hair becomes longer, darker, and extends over the junction of the pubic bones. For pubic hair stage 4, the extent of hair has increased, but has not yet achieved the adult male escutcheon with spread to the medial aspects of the thighs that would be considered Tanner stage 5. Additional features include axillary hair, increased size of the larynx, voice break with deepening of the voice, increased bone mass, and increased muscle strength. Terminal hair develops in androgen-dependent regions on the face and trunk approximately three years after appearance of pubic hair. The distribution and density of beard, chest, abdominal, and back hair varies among individuals.

 

Peak height velocity is both age and sex-dependent. It occurs earlier in girls, between Tanner breast stages 2 and 3, and later in boys, between Tanner testis stages 3 and 4.

 

Approximately 50% of boys experience pubertal gynecomastia (17). Typically, pubertal gynecomastia is transient and most prominent in mid-puberty when the ratio of circulating estradiol to testosterone concentrations is relatively higher.

 

DISCOVERY OF THE HYPOTHALAMIC-PITUITARY-GONADAL (HPG) AXIS

 

Since ancient times, it was known that castration of animals and humans interfered with development of secondary sex characteristics and fertility (14). In 1935, Ernst Laquer and colleagues isolated testosterone from several tons of steer testes (18). Later that year, Adolf Butenandt, Gunter Hanisch, Leopold Ruzicka, and A. Wettstein published the chemical synthesis of testosterone (19, 20). After showing that follicular fluid obtained from a sow ovary was able to induce cornification of vaginal mucosa, Edgar Allen and Edward Doisy isolated the active substance, estrone (21). Donald MacCorquodale, Stanley Thayer, and Edward Doisy isolated estradiol from 8000 pounds of sow ovaries in 1935 (22). Philip Smith, Bernhard Zondek, Hermann Zondek, H.L. Fevold and colleagues, and Geoffrey Harris established the functional relationships involved in HPG axis function (23, 24, 25, 26). Roger Guillemin and Andrew Schally engaged in a vigorous competition to identify hypothalamic releasing hormones including GnRH (27, 28, 29). Ernst Knobil and his colleagues identified that pulsatile GnRH secretion was essential for sustained pituitary gonadotropin secretion (28, 30). Fred Karsch and Ernst Knobil independently developed the concept of the “GnRH pulse generator” (31). In the 1970s, Melvin Grumbach and colleagues measured circulating gonadotropin concentrations in the human fetus (32). Around the same time, Charles Faiman and Jeremy Winter also reported gonadotropin concentrations in normal and agonadal children (33). Their collective findings led to recognition of early postnatal HPG axis activity followed by quiescence of the HPG axis during childhood until resumption of GnRH pulse generator activity at the onset of puberty.

 

Ontogeny of GnRH Neurons

 

Reproductive competence depends on the meticulous development of the GnRH neuron system. In the human fetus, GnRH neurons initially develop in the olfactory placode outside the central nervous system. The olfactory placodes invaginate at approximately 39 days of gestation in the human. Based on the appearance of immunoreactive GnRH protein, the GnRH neuron specification occurs between 39-44 days of gestation (34). The developing GnRH neurons are associated with the embryonic vomeronasal organ. Available data suggest that the GnRH neuron precursors are distinct from those giving rise to the vomeronasal neurons (35).

 

Subsequently, the GnRH neurons migrate accompanied by olfactory-derived axons, olfactory epithelial sheath cells, and blood vessels towards the cribriform plate (36). Migration of the GnRH neurons seems to pause at the nasal/forebrain junction prior to crossing the cribriform plate (37). During this “pause” phase, multiple tissues, chemokines, growth factors, and neurotransmitters appear to form gradients influencing movement of GnRH neurons. Upon reaching the hypothalamus, the GnRH neurons disperse to their final locations sending projections to the median eminence to release GnRH into the hypophyseal portal vasculature.

 

The precise origin and particular factors responsible for the specification and differentiation of GnRH neuron precursors remain enigmatic. Inaccessibility of developing human GnRH neurons has led to development of alternative approaches to elucidate the history of GnRH neurons. One approach has involved a protocol to generate GnRH neurons from human pluripotent stem cells (38). With this approach, Yellapragada et al. demonstrated that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny (39). Using a differentiation trajectory analysis approach, DLX family of transcription factors have been reported to promote in vitro human GnRH neuron differentiation (40).

 

Components of the HPG Axis

 

Gonadotropin-releasing hormone is a decapeptide (pGlu-His-Trp-Ser-Trp-Gly-Leu-Arg-Pro-Gly-NH2) derived from a 92-amino acid precursor, preproGnRH, that was characterized in 1984 (41). LH and FSH are synthesized in the same gonadotroph cell located in the anterior pituitary. LH and FSH are glycoproteins consisting of two subunits. The alpha subunits are identical whereas the beta subunits confer hormone specificity. Each GnRH pulse stimulates an LH pulse.

 

During human gestation, human chorionic gonadotropin (hCG) drives fetal testicular testosterone secretion in the developing male fetus early during gestation. The pituitary gland begins to secrete gonadotropins with LH and FSH becoming detectable in fetal blood after 14 weeks of gestation (42, 43). Initially, pituitary gonadotropin secretion appears to be GnRH-independent with progressive transition to kisspeptin-GnRH regulation of pituitary gonadotropin secretion during the third trimester (44). Peak gonadotropin concentrations occur around the midpoint of gestation followed by a progressive decline attributed to suppression by placental estrogens (45). In the male fetus, testicular testosterone secretion is essential for normal development of internal and external male genital structures. Comparatively, the fetal ovary is quiescent.

 

As noted above, GnRH stimulates pituitary LH and FSH secretion. LH and FSH signal through their cognate receptors which are G-protein coupled receptors (46).

 

GONADS

 

The gonads synthesize sex steroids from cholesterol. In the testis, LH acting through the LH receptor stimulates conversion of cholesterol to testosterone in the Leydig cell. In specific target tissues such as external genital skin and the prostate, testosterone is converted to dihydrotestosterone by the enzyme, 5α-reductase type 2 encoded by the SRD5A2gene. Testosterone influences pituitary LH secretion through negative feedback either via direct actions or indirectly after conversion to estradiol. FSH acting through the FSH receptor promotes growth of seminiferous tubules and supports sperm development. Growth of the seminiferous tubules and increasing numbers of germ cells accounts for increasing testicular volume during puberty.

 

In females, the two cell-two gonadotropin model applies to ovarian steroidogenesis. LH stimulates the theca cell to synthesize androstenedione which diffuses to the granulosa cell where FSH-stimulated aromatase activity stimulates estradiol synthesis. Estradiol has both negative feedback and positive feedback. Estradiol mediated positive feedback is required to elicit the LH surge responsible for ovulation.

 

Activin and inhibin are heterodimeric glycoproteins secreted by the gonads. Inhibins consist of an alpha subunit and one of two homologous yet distinct beta subunits, βA or βB. Inhibin B is composed of an alpha subunit and a βB subunit whereas inhibin A consists of an alpha subunit and a βA subunit. Inhibins are secreted by Sertoli cells in the testes and granulosa cells in the ovary. Inhibin B influences pituitary FSH secretion by negative feedback. In prepubertal boys, inhibin B concentrations reflect Sertoli cell mass and function. After puberty, inhibin B concentrations reflect spermatogenesis (47). Inhibin B correlates inversely with FSH levels in adult men. Activins are dimers of inhibin β subunits, βA, βB and βC; the best characterized are activin A (βAβA) and activin B (βBβB). Activin A stimulates pituitary FSH secretion(48, 49). Follistatin is a monomeric protein that modulates activin activity and can irreversibly inhibit activin activity.

 

Leydig cells secrete insulin-like peptide 3 (INSL3), a small peptide that, in utero, acts through the relaxin-like family peptide receptor 2 (RXFP2) to promote trans-abdominal testicular descent. INSL3 concentrations increase in boys during puberty (50).

 

HYPOTHALAMUS

 

The hypothalamus serves as the rheostat for many physiological functions especially reproduction and growth. The adult human hypothalamus contains approximately 2000 GnRH neurons with cell bodies diffusely distributed in a rostro-caudal continuum (34). The GnRH neurons send projections to the median eminence that terminate in close association with the capillaries of the primary plexus of the hypophyseal portal circulation. Synchronized activity of the GnRH neurons leads to episodic GnRH release into the median eminence with consequent pulsatile pituitary gonadotropin secretion.

 

An extrinsic hypothalamic neuronal network, known as the GnRH pulse generator, governs GnRH neuron function. This network is located within the infundibular nucleus (known as the arcuate nucleus in non-human species). In the human, the GnRH pulse generator is responsible for tonic gonadotropin secretion; pulsatile LH and FSH secretion regulate testicular function in men and modulate ovarian function, especially folliculogenesis in women. In women, the developing follicle secretes increasing amounts of estradiol ultimately triggering an LH surge followed by ovulation. In adult men, pulse frequency is relatively constant at approximately one pulse every 90-120 minutes. Among women, pulse frequency varies across the menstrual cycle from approximately one pulse per hour during the follicular phase and one pulse every 180 minutes during the luteal phase.

 

Among GnRH deficient women, pulsatile GnRH administered at a frequency simulating the follicular phase led to ovulatory menstrual cycles (51). In a preclinical model, administration of pulsatile GnRH to prepubertal rhesus female monkeys initiated pubertal development including ovulatory menstrual cycles (52). Thus, puberty in girls and boys is entirely dependent on resumption of pulsatile GnRH release.

 

Although the GnRH pulse generator was conceptualized by Fred Karsch and Ernst Knobil, the anatomic location of the pulse generator was indeterminant. Identification of loss of function variants in the kisspeptin receptor (KISS1R) gene in patients with congenital hypogonadotropic hypogonadism launched the investigations establishing kisspeptin, neurokinin B, dynorphin, and their cognate receptors as major components of the pulse generator (53, 54). Kisspeptin signals through its receptor, KISS1R, expressed on GnRH cells. Neurokinin B is a decapeptide encoded by the TAC3 (Tac2 in rodents) gene and its cognate receptor encoded by NK3R gene. Both the kisspeptin and neurokinin B receptors are G-protein coupled receptors. Dynorphin is an opioid peptide that signals through a kappa-opioid receptor which is also a G-protein coupled receptor.

 

Due to the inaccessibility of human brain, especially the pubertal brain, the contemporary model of the GnRH pulse generator has been delineated by preclinical studies performed in rodents, sheep, and non-human primates (55). This model predicts that reciprocal interactions within a network of kisspeptin neurons in the infundibular nucleus leads to synchronous intermittent activation transmitted to GnRH neurons by kisspeptin fibers that project to the median eminence. These kisspeptin fibers are closely associated with GnRH projections targeting the portal capillaries (56).

 

Based on the detection of kisspeptin, neurokinin B, and dynorphin in the arcuate kisspeptin neurons of mice and sheep, these neurons have been labeled as KNDy neurons (57). Preclinical data suggest that KNDy neurons serve as the intrinsic GnRH pulse generator (58). Kisspeptin and neurokinin B stimulate GnRH release whereas dynorphin appears to be inhibitory. Coordinated interactions of these neuropeptides within the arcuate kisspeptin neuronal network are ostensibly central to the neurobiology of the GnRH pulse generator resulting in pulsatile kisspeptin output. However, the applicability of these findings to human biology remains to be confirmed.

 

In humans, the HPG axis is active during gestation and the early neonatal period followed by the quiescent years of childhood until the onset of puberty occurs. This pattern suggests that diverse mechanisms integrate the hierarchical activation and deactivation of various stimulatory and inhibitory neuronal pathways ultimately regulating pubertal onset and progression towards reproductive maturity. Thus, a central inhibition of the axis occurs during childhood. For puberty to occur, increased expression of the key factors, KISS1, NKB3, and GnRH, must begin along with decreased expression of the various inhibitory factors. In other words, during the pubertal transition, the balance between inhibitory and stimulatory factors shifts to favor the re-activation of the HPG axis, onset of pubertal changes, and reproductive competence.

 

Identifying the proximate factors and specific interactions responsible for the on-off-on pattern of HPG axis activity in humans has been a longstanding enigma. Starting with clinical findings, the availability of more sophisticated tools and preclinical models have begun to identify pieces of the puzzle to elucidate the fine details of HPG axis functioning. One factor involved in the suppression of puberty was identified in families with paternally inherited GnRH-dependent/central precocious puberty (CPP). Exome sequencing analyses in multiple families with CPP identified loss of function variants in the makorin 3 (MKRN3) gene (59). This gene, mapped to the Prader Willi region at chromosome 15q11.2, is exclusively expressed from the paternal allele. Consistent with the hypothesis that MKRN3 suppresses the GnRH pulse generator, circulating MKRN3 concentrations decline during puberty (60, 61, 62). 

 

The MKRN3 protein is an E3 ubiquitin ligase consisting of 507 amino acids. It is expressed in KNDy neurons. The protein has five zinc finger domains. Regarding its function, the protein can ubiquitinate substrates and can undergo auto-ubiquitination (63). MKRN3 ubiquitinates methyl-CpG-DNA binding protein 3 (MBP3) interfering with GnRH1transcription (64). Available preclinical data suggest that MKRN3 functions as a brake on neuronal GnRH release (65). One potential factor influencing MKRN3 expression is microRNA (miRNA) miR-30. Using a rat model, hypothalamic miR-30 expression increased while Mkrn3 expression decreased during puberty. In addition, treatment with agents that interfered with the binding of miR-30 to Mkrn3 were associated with delayed puberty in female rats (66). Using proteomics, MKRN3 targets include insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and several members of the polyadenylate-binding protein family (67). The decline of hypothalamic Mkrn3 expression in mice and serum MKRN3 protein levels in females prior to the onset of puberty support the hypothesis that MKRN3 suppresses pubertal initiation possibly through effects on prepubertal hypothalamic development and plasticity (61, 67)

 

Preclinical studies have provided persuasive evidence regarding the regulatory role of epigenetic modifications in pubertal maturation. Epigenetics refers to changes in gene expression and/or activity independent of changes in the primary nucleotide sequence (68). Epigenetic changes include DNA modifications such as methylation/demethylation and histone post-translational modifications such as acetylation/deacetylation. Other post-translational protein modifications such as ubiquitination may also influence protein function. Ubiquitination involves the transfer of ubiquitin to a protein altering its function typically by interfering with protein actions or by promoting protein degradation. As noted above, the MKRN3 protein can function as a ubiquitin ligase. Noncoding RNAs such as miRNAs provide yet another regulatory mechanism.

 

Another example of epigenetic regulation of pubertal maturation involves two mutually antagonistic histone methylating complexes, the Poly-comb and Trithorax groups. The Poly-comb group represses gene transcription while the Trithorax group appears to function as a gene activator. Preclinical studies performed in rats showed that the Poly-comb group effectively silenced Kiss1 expression until the onset of puberty when increased methylation of the Eed and Cbx7 genes occurred leading to decreased Eed and Cbx7 expression and increased Kiss1 expression (69). Recruitment of the Trithorax activity group further enhanced.  Kiss1 expression (70, 71). Genome wide association studies have implicated zinc finger (ZNF) genes. In nonhuman primates, expression of two ZNFs, GATAD1 and ZNF573, decreases upon pubertal reactivation of the GnRH pulse generator (71).

 

Clinically, it has long been recognized that extremes of body energy status such as chronic malnutrition or severe obesity influence the HPG axis especially in girls and women. The hypothalamic kisspeptin neurons integrate various peripheral and central metabolic signals reflecting energy intake, energy expenditure, and environmental circumstances. Signal coordination between reproductive and metabolic neurons can be direct or indirect. For example, leptin does not directly regulate kisspeptin neurons yet acts as a permissive factor for the onset of puberty (72). Cellular energy and metabolic sensors include mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and sirtuin 1 (SIRT1) (73). Depending on energy status, mTOR and AMPK promote or repress puberty, respectively, by activating or inhibiting Kiss 1 neurons in the arcuate nucleus. Other factors such as melanocortin and agouti-related peptides also interact with kisspeptin pathway (74). In the hypothalamus, neuronal nitric oxide (NO) appears to act on GnRH neurons to integrate metabolic and gonadal information (75, 76). Detailed reviews regarding the neurobiology of the GnRH pulse generator are beyond the scope of this chapter and are available elsewhere (77, 78, 79, 80, 81, 82).

 

MINI-PUBERTY

 

Facilitated by the availability of more sensitive hormone assays, Forest and her colleagues described a transient period of increased HPG axis activity in early infancy (83, 84). Following the low gonadotropin concentrations at birth, gonadotropin concentrations were found to rise in both boys and girls within weeks of birth (85). This period of transient gonadotropin secretion has been designated as “minipuberty”. Gonadotropin concentrations in the immediate neonatal period are likely low due to in-utero suppression by placental estrogen. With removal of the placental estrogen suppression, the HPG axis is functional. Relevantly, physical findings typical of pubertal sex steroid secretion are absent with the rare exception of vaginal bleeding attributed to decreased exposure to placental estrogen.

 

Over the first few years of life, sexual dimorphism in gonadotropin concentrations occurs (86) Boys have higher LH concentrations which peak between 2-10 weeks of age and decline by 4-6 months of age. Girls have higher FSH concentrations which may remain elevated until 2-4 years of age.

 

In boys, LH stimulates testicular testosterone secretion with testosterone concentrations typically peaking around 1 month of age followed by a decline to prepubertal concentrations by 7-12 months of age. During this phase, the number of germ cells and Sertoli cells increase and penile size increases (87, 88). The proliferation of Sertoli cells leads to a transient increase in testicular volume (89). Sertoli cells secrete Anti-Mullerian Hormone (AMH) and inhibin B. Since Sertoli cells do not express androgen receptors during this stage, spermatogenesis does not occur and AMH secretion remains high (90, 91). A temporary increase in the number of Leydig cells also occurs, but subsequent fetal Leydig cell apoptosis reduces fetal Leydig cell number (92). Longitudinal data obtained from healthy boys suggests a temporal dissociation of Leydig and Sertoli cell activity during minipuberty (93). These data suggest that single blood sample may be insufficient to assess HPG axis activity during early infancy and that obtaining several consecutive samples may be more informative. Curiously, gonadotropin and testosterone concentrations are higher among preterm boys. In addition, increases in testicular volume and penile length are greater in preterm boys compared to full term boys essentially enabling catch-up for testicular volume and penile length (94). Some small studies have documented an exaggerated physiologic hormonal response in extremely premature infants (95).

 

In girls, the gonadotropins promote granulosa cell proliferation and ovarian estrogen and AMH secretion (96). As would be anticipated, AMH concentrations remain much lower in girls compared to boys (97). A longitudinal study involving healthy full-term infant girls demonstrated two gonadotropin peaks in early infancy with one peak occurring around days 15 to 27 and a later peak occurring at days 164-165 (98). Again, collecting several consecutive samples may be more informative than a single blood sample to assess for minipuberty in infancy.

 

This transient time period of an active HPG axis, provides an opportunity to diagnose individuals with differences/variants of sex development (DSD/VSD). In a series including both healthy infants and infants with DSD, testosterone measured by LC-MS/MS, AMH concentration, and LH/FSH ratio provided the best discrimination between sexes. The cut-point for LH/FSH ratio was 0.32. Inhibin B and AMH levels were higher in boys with minimal overlap in girls (99). Infants with Turner Syndrome usually have elevated FSH concentrations. Surprisingly, gonadotropin concentrations are typically not elevated in patients with complete androgen insensitivity.

 

This brief interval of HPG axis activity can also help diagnose congenital hypogonadotropic hypogonadism in boys who present with micropenis accompanied by low gonadotropin and testosterone concentrations (100). Testosterone, LH, FSH, AMH, and inhibin B concentrations may provide information regarding the functionality of testicular tissue in infant boys (101).

 

As noted above, the human HPG axis displays an “on-off-on” pattern. The biological basis and rationale for transient post-natal HPG axis activity during the first few months of life are enigmatic. At birth, the brain is still plastic with ongoing development. Most axon and synapse formations are completed during the first year of life. Does this transient HPG axis activity imprint specific areas in the brain? Does minipuberty influence future patterns for female and male reproductive function with cyclic gonadotropin patterns in females and not in males? Are gonadal hormones during infancy able to affect future fertility, gender identity, sexual orientation, behaviors, and risk for autism spectrum dysfunction? Data are accruing regarding patterns of hormone secretion during the first six months of life. However, the factors that initiate and terminate this transient period of HPG axis activity and maintain the quiescence of the HPG axis until the onset of puberty are still unknown.

 

 

Over the past few decades, several studies have observed that puberty is beginning at a younger age. Clinical studies examining ages of the onset of puberty depend on the criteria used to denote puberty. Onset of breast development and age at menarche are the conventional indicators of puberty in girls. Prospective observations and retrospective questioning of parents and young girls through in-person questioning has been used to record age at menarche; shorter recall intervals provide the greatest accuracy regarding the details of menarche (102, 103). For boys, age at voice change has been used as a surrogate marker because accurate ascertainment of pubertal onset in boys requires testicular exams using an orchidometer, thus, effectively excluding large-scale epidemiologic clinical studies (104).

 

During medieval times, available evidence suggests that puberty began around 10-12 years of age. However, the tempo of puberty was slow with menarche occurring closer to 15 years in rural areas and 17 years in London (105). Presumably, undernutrition, increased infections, and greater physical exertion impacted both the timing and tempo of puberty during medieval times (106). The age of menarche declined from 16 to 17 years in the early 19th century to 13 years of age in the late 20th century in Europe and North America. Similarly, the age at menarche has declined in the Yunnan Province in China (107). This decline has been attributed to the improvement in socioeconomic conditions. Currently, the dialogue continues as to whether the trend towards earlier puberty is persisting and, if so, what are the factors driving this process. 

 

Data regarding pubertal milestones in American girls were obtained through the cross-sectional Third National Health and Nutrition Examination Survey (NHANES III) between 1988 and 1994.  Among these American girls, mean ages in years for breast development, pubic hair development, and menarche were 9.5, 9.5, and 12.1 for non-Hispanic black girls; 9.8, 10.3, and 12.2 for Mexican-American girls; and 10.3, 10.5, and 12.7 years for non-Hispanic white girls, respectively (108). In 1997, the Pediatric Research in Office Settings (PROS) study reported earlier onset of thelarche with the caveat that breast palpation was not performed (109). The Copenhagen Puberty Study reported that mean age at breast development was lower in the 2006 cohort compared to the 1991 cohort whereas mean age at menarche was similar in both cohorts. Independent of BMI, gonadotropin concentrations were comparable between these cohorts while estradiol concentrations were lower in the 2006 cohort (110).

 

Beginning in 2004, the Breast Cancer and Environment Research Program (BCERP) prospectively recruited three cohorts of girls aged 6-8 years. This program recruited non-Hispanic white, Hispanic, non-Hispanic black girls, and Asian girls living in New York, Ohio, and California. The overall median age at menarche was 12.25 years with ethnic background median ages as follows: Hispanic girls 11.6 years, black girls at 11.8 years, white girls at 12.5 years, and Asian girls at 12.0 years (111). This cohort differed from the NHANES III study because Hispanic girls experienced menarche earlier than the black girls. These studies, all performed in the United States, report race and ethnicity-related differences in onset of pubertal milestones. Detailed assessment of the potential impact of socio-economic factors was not performed. Notably, differences noted in pubertal timing are smaller than the overall variation among individuals in the population. Most importantly, clinical decision-making should reflect an individual patient's characteristics and family history with less dependence on racial or ethnic backgrounds.

 

Comparable studies from Spain and Greece have also reported earlier onset of breast development and slower pubertal tempo (112, 113). Thus, available data including a systemic review of international studies largely confirm the ongoing trend for earlier breast development with minimal decline in age at menarche (114).

 

Several questions regarding this earlier onset of puberty, predominantly earlier thelarche, need to be considered. Does this earlier breast development reflect earlier resumption of GnRH pulse generator activity, extragonadal estrogen production, or environmental exposures? What, if any, is the relationship of BMI to puberty? Another consideration is that race/ethnicity are socio-political constructs and are not fully representative of biology. While genetic ancestry likely influences the onset of puberty, nutritional factors and environmental exposures play important roles. Hence, should cut-off points based primarily on race/ethnicity continue to be utilized?

 

Based on single unstimulated gonadotropin concentrations, data from the Copenhagen puberty in girls study suggest that gonadotropin concentrations are not obviously increased in girls with early thelarche. Thus, the phenomenon of early thelarche appears to be independent of gonadotropin secretion and may not signify early resumption of GnRH pulse generator activity (115).

 

The possibility that exposure to endocrine-disrupting chemicals (EDCs) can induce early thelarche has been questioned. EDCs are defined as exogenous chemicals that interfere with hormone action. EDCs include phthalates, phenols, phytoestrogens, organochlorine pesticides, polybrominated flame retardants, diphenyl ethers, heavy metals, and perfluorochemicals. In addition to pesticides, these chemicals can be found in common household products such as hair products, soaps, toothpaste, perfumes, plastics, essential oils, and cleaning products (116). Valid assessment of the consequences of EDCs on puberty is problematic because exposure may occur in utero and generally involves a mixture of assorted EDCs with differing half-lives and activities. Differences in the duration and route of the exposure(s), methodology to detect EDCs, and potential sample contamination further confound analyses. One potential example regarding EDCs involved transient past exposure to organochlorine pesticides among internationally adopted girls in Belgium who subsequently developed precocious puberty (117). Animal models suggest that EDCs can affect puberty through epigenetic mechanisms (118). Nevertheless, most data available regarding the consequences of EDCs on human puberty are inconclusive (119).

 

Relationship with BMI

 

Observational data has shown a relationship between BMI and age at puberty in girls (120, 121). The BCERP study found that girls who were overweight or obese at baseline experienced menarche 0.3 years earlier with age at thelarche being inversely correlated with BMI. The BCERP also concluded that BMI had a greater effect than ethnic background on age at menarche (111). Limited data exist regarding the relationship of BMI to pubertal onset in boys. The Puberty Cohort of the Danish National Birth Cohort reported that increased BMI was associated with earlier onset of puberty in boys and girls (122). Among boys, pubertal milestones, testicular enlargement, voice break, and testosterone concentrations showed inverse correlation with BMI (104). Hence, available evidence strongly indicates an inverse relationship between BMI and the onset of puberty in both boys and girls.  

 

Yet, investigating the relationship between puberty and BMI is confounded by potential hormonal and genetic influences (123). Obesity may be associated with hyperinsulinemia and lower sex hormone binding globulin concentrations with consequent higher free sex steroid concentrations. In addition, some genes influence both BMI and pubertal timing (124, 125). The pro-opiomelanocortin (POMC) and central melanocortin systems provide one example of the intricate interrelationships between nutrient signaling and reproductive function. Neurons expressing POMC, producing α-MSH (melanocyte-stimulating hormone), have been suggested to stimulate puberty onset and gonadotropin secretion via modulation of arcuate Kiss1 neurons (126, 127).

 

Genetic Factors

 

Genetic  factors influence pubertal timing as evidenced by twin studies demonstrating > 50% hereditability for menarche (128). Skeletal maturation, age at pubertal growth spurt, and Tanner staging also show greater concordance between monozygotic twins compared to dizygotic twins emphasizing the relevance of genetic variation in the timing of puberty. Thus, 50-80% of variation in the timing of puberty onset may reflect genetic variation (129). Parental self-reports regarding pubertal timing are associated with timing of specific pubertal milestones in offspring of the concordant sex (130, 131). Genome-wide association studies (GWAS) have detected loci associated with age at menarche (132). Some loci appear to be common and independent of ancestry. A large-scale trans-ethnic GWAS, involving 38,546 women of diverse and predominantly non-European ancestry or ethnicity, identified a novel locus in chromosome 10p15 that is associated with early menarche. This region maps to intron 7 of the aldo-keto reductase Family 1, member C4 (AKR1C4) gene, a member of family of enzymes involved in steroid metabolism and action (133).

 

To summarize, the secular trends suggesting an earlier onset of puberty appear to be persistent although the age at menarche appears to be relatively static. Likely contributing factors include the rising prevalence of obesity, exposure to potential EDCs, specific dietary influences, and decreased physical activity.

 

VARIATIONS IN PUBERTAL DEVELOPMENT

 

Timing of the onset of puberty reflects complex interactions between hormonal and neuronal signals with genetic, metabolic, and environmental factors. These interactions presumably begin early in development and ultimately lead to the re-activation of the HPG axis concomitant with the onset of puberty. Multiple factors, both known and unknown, influence the reactivation of the GnRH pulse generator modulating pubertal onset. As noted above, familial patterns of pubertal development and twin studies highlight the role of genetic factors. Studies of families with either delayed or precocious puberty led to discovery of genes relevant to pubertal onset. In addition, genetic factors including single nucleotide polymorphisms (SNPs) have been associated with pubertal timing in both sexes and across ethnic groups. Epigenetic mechanisms have been suggested to affect the development and function of the GnRH neuronal network ultimately influencing HPG axis function. How confounders such as socioeconomic, environmental, and nutritional status influence pubertal development is unclear. These factors can influence puberty timing, HPG axis function, and fertility.

 

Precocious puberty is defined as the development of puberty prior to age 8 in girls, and age 9 in boys (134, 135). In girls, delayed puberty is defined as the absence of breast development by age 13 years, absence of menarche by age 15 or lack of menses after 3 years since breast development. In boys, delayed puberty is defined as absence of pubertal development by age 14 (136). Evaluation of a child with abnormal timing of puberty entails thorough knowledge of normal pubertal development, typical variations of normal pubertal development, and causes of abnormal pubertal development. The next section focuses on the evaluation of a patient presenting with a variation in pubertal development.

 

PRECOCIOUS PUBERTY  

 

Traditionally, the diagnosis of precocious puberty is considered when signs of puberty develop prior to 8 years of age in girls and 9 years in boys (137). These ages are based on Tanner’s original observations on English children regarding typical ages at specific pubertal stages. However, these age criteria should be used as guidelines to complement the evaluation of individual patients. Precocious puberty can be categorized as central or gonadotropin-dependent precocious puberty (CPP) or non-gonadotropin-dependent or peripheral precocious puberty (PPP). Additionally precocious puberty can be further classified as familial or sporadic and syndromic or non-syndromic. The specific etiologies and management differ between the two broad categories of CPP or PPP. Potential consequences of early puberty and menarche in girls include increased risks for breast cancer and diabetes as adults (138, 139).

 

Central Precocious Puberty or Gonadotrophin Dependent Precocious Puberty

 

Central precocious puberty (CPP) is associated with early maturation of the HPG with premature reactivation of the GnRH pulse generator and sequential maturation of breasts and pubic hair in females. In males, sequential maturation of testicular volume, penile enlargement, and pubic hair is observed. Typically, the pubertal characteristics are appropriate for the child's sex (isosexual). Despite the earlier onset of puberty, the sequence of pubertal events is usually normal. CPP is due to organic lesions in approximately 40-100 percent of boys whereas idiopathic precocious puberty is the most common diagnosis in girls (69-98%) (140). These children have accelerated linear growth for age, advanced bone age, and pubertal levels of LH and FSH. A Spanish observational report described an annual incidence of CPP ranging between 0.02 and 1.07 new cases per 100,000 (141) while a Korean study reported an incidence of 15.3 per 100,000 girls, and 0.6 per 100,000 boys (142). Distinguishing among CPP, isolated premature thelarche, and premature adrenarche is important because the pathophysiology and therapeutic interventions differ.

 

CNS LESIONS/INSULTS

 

CPP can be associated with central nervous system lesions. Hamartomas of the tuber cinereum are congenital benign lesions comprised of heterotopic gray matter, neurons, and glial cells. The prevalence is approximately 1 in 200,000 children (143). Hamartomas are the most commonly recognized CNS lesions associated with CPP in very young children. Hamartomas can be categorized as para-hypothalamic, attached or suspended from the floor of the third ventricle, or as intrahypothalamic, in which the mass is enveloped by the hypothalamus and distorts the third ventricle. The lesions do not grow over time, do not metastasize, and do not produce β-human chorionic gonadotropin-(β-hCG). In some instances, hamartomas are associated with gelastic (laughing or crying) seizures. Yet, most patients with hypothalamic hamartomas do not display neurological symptoms (144, 145). Most hypothalamic hamartomas are sporadic and appear to be idiopathic. Hypothalamic hamartomas can also occur in Pallister-Hall Syndrome (PHS) and oral-facial-digital syndrome (OFD) types I and VI (146). Genetic variants in the sonic hedgehog pathway have been associated with hypothalamic hamartoma (147, 148). The mechanism(s) through which hypothalamic hamartomas lead to CPP is unknown. Hamartoma located close to the infundibulum or tuber cinereum are often associated with CPP whereas those functionally connected to the mammillary bodies and limbic circuit are typically associated with epilepsy without CPP (149, 150). As discussed below, medical treatment is usually indicated for hypothalamic hamartomas associated with CPP. Surgical treatment should be limited to large hamartomas complicated by severe refractory drug-resistant epilepsy (151).

 

CNS tumors such as astrocytomas, ependymomas, and pinealomas have rarely been associated with CPP. Among girls, factors associated with CNS lesions include: (1) age younger than 6 years; (2) absence of pubic hair; and (3) estradiol concentrations greater than 30 pg/ml (110 pmol/L) (152, 153). As noted above, suspicion for CNS lesions is higher for boys than for girls.

 

Neurofibromatosis type 1 (NF1) is an autosomal dominant multi-system neurocutaneous disorder due to loss-of-function variants in the neurofibromin-1 (NF1) gene located at chromosome 17q11.2. NF1 is often associated with CPP typically due to optic glioma. The glioma is usually a benign pilocytic astrocytoma that can occur anywhere along the optic tract; the most common locations are within the optic nerve or chiasm. CPP has also been described in NF1 in the absence of optic glioma (154). Children with meningomyelocele and spina bifida also have an increased incidence of CPP. Although the precise mechanism responsible for CPP in these children is unclear, associated factors may include increased perinatal intracranial pressure and brainstem malformations such as Chiari II malformations (155). The mechanistic link between CPP and Rathke cleft cysts, Chiari malformation, and pineal and arachnoid cysts is unclear.

 

Septo-optic dysplasia (SOD) is a heterogeneous congenital condition characterized by presence of at least two features of the classic triad which include optic nerve hypoplasia, anterior pituitary hormone deficiencies, and midline brain anomalies. SOD is associated with genetic variants in HESX1, SOX2, SOX3, and OTX2 genes. Although SOD is typically associated with delayed puberty, CPP can occur (156, 157).

 

CNS tumors may be treated with CNS irradiation (158). In some instances, CNS irradiation is associated with acquired CPP (159). In this situation, concurrent growth hormone (GH) deficiency may be present. The linear growth spurt of CPP may mask the decreased linear growth velocity due to GH deficiency. Hence, in this setting, consideration should be given to evaluating the GH axis by provocative GH testing. If testing shows GH deficiency, the patient may benefit from treatment with GH combined with GnRH agonist therapy. Rarely, CPP occurs following head trauma and can develop many years after the injury(160, 161).

 

SECONDARY CPP

 

Some children exposed to elevated circulating high sex steroid concentrations occurring in other disorders such as McCune-Albright syndrome, congenital adrenal hyperplasia, and virilizing adrenocortical tumors may develop a secondary CPP (163). These individuals typically have accelerated bone age maturation. The precise mechanism responsible for development of the secondary CPP is unclear. The secondary CPP may represent a priming effect of sex steroids on the hypothalamus or potentially as the consequence of the acute decrease in sex steroid concentrations with treatment of the underlying etiology (164) (165).

 

NON-SYNDROMIC CPP

 

Specific genetic variants have been associated with non-syndromic CPP (See Table 1) (166). Loss of function MKRN3variants are the most reported genetic cause of familial CPP. Paternally inherited loss-of-function MKRN3 variants have been reported in up to 33-46 percent of familial cases of CPP and nearly 0-20% percent of sporadic cases (167) . To date, at least 70 deleterious MKRN3 variants have been identified in patients with CPP. These variants lead to diminished inhibition of puberty results in early onset of puberty. Differing ubiquitination patterns suggests that MKRN3 has multiple molecular mechanisms associated with CPP (168). Curiously, a GWAS study investigating parental effects on pubertal development reported that the paternal allele of a specific SNP (rs12148769, G>A) in MKRN3 was associated with age at menarche in healthy girls suggesting that variants in this region affect pubertal timing within the normal range (132). Although circulating MKRN3 concentrations decrease with onset of puberty, peripheral blood MKRN3 concentrations are not adequately sensitive to distinguish CPP (169).

 

TABLE 1. Genes Associated with Central Precocious Puberty (175, 461)

 

Gene

(Reference/s)

Protein encoded

Genetic locus

Comments

MKRN3

(59, 63, 167, 462)

Makorin ring finger protein 3

15q11-q13

Loss-of-function mutation

KISS1R (previously named GPR54)

(463, 464, 465)

Kisspeptin receptor

19p13.3

Gain-of-function mutation

KISS1

(465)

 

Kisspeptin

1q32

Gain-of-function mutation

DLK1

(466, 467, 468)

Delta-like homolog 1

14q32

-Loss-of-function mutation

-Metabolic abnormalities (obesity, type 2 diabetes, hyperlipidemia)

ESR1

(469, 470)

Estrogen receptor 1

6q25.1-q25.2

Mutations/polymorphisms, epigenetic change

CYP19A1

(471)

 

Aromatase

15q21

(TTTA)n polymorphism, epigenetic change

 

Evaluation of another family with CPP led to identification of a loss of function variant in the delta-like 1 homologue (DLK1) gene. DLK1, also known as preadipocyte factor 1, plays a role in the Notch signaling pathway. DLK1 is a paternally expressed gene located at chromosome 14q32.2. Two differentially methylated regions influence the DLK1 imprinting pattern. DLK is located within the genetic locus associated with Temple syndrome. Temple syndrome is characterized by prenatal growth retardation, hypotonia in infancy, motor delay, small hands, CPP, and short stature. In addition to DLK1 loss, two other genes from the paternally inherited chromosome, RTL1 and DIO3, results in Temple Syndrome. Genetic findings associated with Temple syndrome include maternal uniparental disomy, paternal deletion, or loss of differential methylation at the DLK1/MEG3 region on chromosome 14 (170). Women with DLK1 variants also have a metabolic phenotype characterized by overweight/obesity and insulin resistance (171).

 

Gain-of-function variants in the kisspeptin 1 gene (KISS1) and its cognate receptor, KISS1R, gene have been identified in children with CPP. A heterozygous variant in the KISS1 gene, p.Pro74Ser, was identified in a boy who developed CPP at one year of age; in vitro studies suggested that this variant was more stable than the normal protein leading to a prolonged duration of action (172). A girl with precocious puberty was found to have a variant in the KISS1R gene; in vitro studies of this p.Arg386Pro variant showed prolonged activation of the intracellular signaling pathways following kisspeptin stimulation (173, 174).

 

Among a series of 586 children with familial CPP, both maternal and paternal inheritance patterns were found. Variants in MKRN3 were the most common cause in paternally inherited CPP. Among the maternally inherited cases, genetic analysis detected rare variants of unknown significance (175).

 

SYNDROMIC CPP

 

In addition to genetic and idiopathic CPP, CPP can occur as a feature in specific syndromes. Pallister-Hall and Temple Syndrome are described above. Other syndromes associated with CPP include Cowden and Cowden-like cancer predisposition syndromes associated with PTEN, SDHB-D and KLLN gene variants. These disorders are characterized by multiple multisystemic hamartomas which may be associated with CPP when the skull base, infundibulum, or hypothalamus are affected. Although Prader-Willi syndrome is typically associated with delayed puberty, CPP has also been reported (176). Other genetic syndromes associated with CPP include tuberous sclerosis and Williams-Beuren (See Table 2). Williams-Beuren is associated with genetic variant at chromosome 7q11.23 (177). Rare cases of precocious puberty have also been reported in Russell Silver syndrome (178).

 

Table 2. Syndromic Causes of Central Precocious Puberty Without CNS Lesions (CPP)

Gene (Reference/s)

Genetic locus

Comments

MECP2

methyl-CpG-binding protein 2

(472)

 

Xq28

Rare forms of Rett syndrome

X-linked dead-box helicase 3

(461)

Xp11.4

Neurodevelopmental delay

Xp22.33 deletion, SHOX region

(473)

Xp22.33

Body disproportion, short stature, Madelung deformity

Xp11.23-p.11.22 duplication

(474)

Xp11.23-p11.22

Intellectual disability, speech delay, electroencephalogram abnormalities, excessive weight, skeletal anomalies

Temple syndrome

-DLK1

Maternal uniparental disomy or paternal deletion

(170, 473)

14q32.2

Imprinting defect, act via DLK1,

Prenatal and postnatal growth failure, hypotonia, small hands and/or feet, obesity, motor delay

Prader-Willi syndrome

- MKRN3

Paternal deletion or maternal uniparental disomy of chromosome 15q11-q13

(475)

15q11-q13

Changes to the imprinted MKRN3 and/or MAGEL2genes

Hypotonia, obesity, growth failure, cognitive disabilities, hypogonadism

Silver-Russell syndrome

Hypomethylation of chromosome 11p15 or maternal uniparental disomy of chromosome 7       

(476)

 

11p15.5

Possible imprinted or recessive factors, not well elucidated,

Prenatal and postnatal growth retardation, relative macrocephaly, prominent forehead, body asymmetry, feeding difficulties

Williams-Beuren

(177, 477, 478)

7q11.23

Distinct face, cardiovascular disease, short stature, intellectual disability, hyper-sociability

Kabuki syndrome

(479)

12q13.12

Downregulation of estrogen receptor activation

Neurodevelopmental phenotypes, typical distinct face, short stature

Mucopolysaccharidosis type IIIA or Sanfilippo disease

(480)

17q25.3

Severe neurologic deterioration, visceromegaly, skeletal abnormalities

 

NONPROGRESSIVE PRECOCIOUS GONADARCHE

 

Some children experience a nonprogressive (or slowly progressing) CPP (179). Typically, basal gonadotropin concentrations are prepubertal. In general, children with nonprogressive CPP show no or minimal pubertal responsiveness to GnRH stimulation. Height potential is generally unaffected. Typically, these individuals do not usually benefit from GnRH-Ra therapy. Physical findings alone cannot distinguish between progressive and nonprogressive CPP. Presumably this early pubertal development reflects a transient premature activation of the GnRH pulse generator. Longitudinal follow-up to assure that puberty is not progressive is the most appropriate management.

 

GONADOTROPH ADENOMA

 

The anterior pituitary gland consists of highly differentiated ectoderm-derived cells expressing specific hormones such as LH, FSH, GH, prolactin, and ACTH. LH and FSH are secreted by gonadotrophs which are derived from the steroidogenesis factor 1(SF-1) lineage. Gonadotroph adenomas, a type of pituitary adenoma, account for approximately 40% of pituitary adenomas  (180, 181) in adults. In children, gonadotroph adenomas can very rarely cause central precocious puberty (182). Though, most gonadotroph adenomas are nonfunctional and benign, rare cases of functional adenomas have been reported. Hormone profiles of functioning adenomas most commonly show elevated FSH concentrations with or without increase in LH concentrations. Elevated TSH secretion resulting in hyperthyroidism may occur concurrently (180, 181).

 

GUT MICROBIOME AND PUBERTY

 

Microbiota interact with a variety of metabolic and endocrine pathways of the host through genetic expression of more than 100 times the human genome. The gut microbiome variety, composition and impact on health depend on a vast number of variables, both internal, such as age, genetic factors, gender, and endocrine and immune systems, as well as external factors, such as diet, environment, drugs, and pathogens. The relationship between sex hormones and gut microbiome is complex. Sex steroids may directly or indirectly influence the sex-specific gut microbiome that develops during puberty (183). One study reported several gut microbiome alterations in girls with CPP including Ruminococcus bromii, Ruminococcus callidus, Roseburia inulinivorans, Coprococcus eutactus, Clostridium sporosphaeroides, Clostridium lactatifermentans, Alistipes, Klebsiella and Sutterella (176). Although the evidence of the interaction between microbiota and sex hormones remains limited, evidence of diversity of the gut microbiota at different pubertal stages and that alterations may occur in girls with CPP represents an area for potential future development in the prediction and prevention of precocious puberty (184).

 

Treatment of central precocious puberty

 

GONADOTROPIN-RELEASING HORMONE ANALOGS

 

Long-acting Gonadotropin-releasing hormone analogs (GnRHa) have been the standard treatment of CPP since the mid-1980s (185, 186). The GnRHa are super-agonists that bind to the pituitary GnRH receptor downregulating the endogenous pituitary GnRH receptor resulting in decreased gonadotropin and sex steroid secretion. These medications are modified preparations of the native GnRH decapeptide engineered to increase potency and duration of action by substituting a D-isomer amino acid for the naturally occurring L-glycine at position 6. In some analogs, the tenth amino acid is deleted with modification of the naturally occurring L-proline at position 9 (14).

 

Several distinct GnRHa preparations are available differing in route of administration and duration of action (See Table 3) (28). The choice of a specific GnRHa depends on patient, caregiver, and physician preference and on insurance coverage/payment/authorization. Treatment with GnRHa leads to regression or stabilization of pubertal symptoms, deceleration of linear growth velocity, and slowing of skeletal maturation. Some girls experience estrogen withdrawal bleeding about 2-3 weeks following the first injection. Parents and the patient should be counseled to expect this episode of vaginal bleeding (187).  

 

 Table 3. Currently Available GnRHa Therapeutic Options

GnRHa Preparations                         

Dose  

Frequency       

Route

Goserelin

3.6mg

Once a month

intramuscular

Leuprolide

7.5mg

Once a month

intramuscular

 

11.25mg

Once a month

intramuscular

 

15mg

Once a month

intramuscular

 

11.25mg

Every 3 months

intramuscular

 

30mg

Every 3 months

intramuscular

 

45mg

Every 6 months

intramuscular

Leuprolide

45mg

Every 6 months

subcutaneous

Triptorelin

22.5mg

Every 6 months

intramuscular

Nafarelin

800mcg

Twice daily

intranasal

Histrelin

50mg

Annually *

Subdermal implant

*May be used up to 2 years (481).

 

Adverse Effects

 

In general, GnRHas are safe and effective. Adverse events include injection site reactions and sterile abscesses at the site of the injection or implant (188, 189, 190) which may result in loss of efficacy. Minor reported side effects include headaches, hot flashes, vaginal withdrawal bleeding, and mood swings (191). Extremely rare side effects include hypersensitivity reactions, seizures, slipped capital femoral epiphysis, idiopathic intracranial hypertension, and anaphylaxis. One concern regarding the histrelin implant is possible device fracture during extraction; ultrasound-guided removal of the remaining fragments may be necessary (192).

 

GnRHas, specifically only leuprolide and degarelix, have been associated with prolonged QT interval. A prolonged QT interval increases the risk of developing torsades de pointes (TdP) which is a ventricular arrhythmia associated with sudden cardiac death. Low serum potassium or magnesium may exacerbate the risk for prolonged QT interval. Individuals also taking anti-psychotics (typical and atypical), anxiolytics, and anti-depressants may have an increased risk for prolonged QT intervals when taking leuprolide. Hence, providers should inquire regarding other medications, history of congenital heart disease, and family history of Long QT Syndrome or sudden death. If positive, the provider should obtain screening and follow-up EKGs.

 

Studies conflict regarding how GnRHa treatment impacts weight gain and BMI. Some studies have reported weight gain during treatment (193, 194, 195, 196) whereas others have not found any significant change in weight or BMI (197, 198).As with all patients, counseling patients regarding the pre-treatment weight trajectory and healthy lifestyle is beneficial. Women with a history of CPP have been reported to have similar adult weight to the general population (199).

 

Bone mineral density is typically elevated at diagnosis with deceleration in bone mineral accrual during treatment. However, follow-up several years after treatment shows normal bone mineral density compared to population norms (200). Available outcome data suggest that fertility is not compromised for women or men with histories of CPP (192, 201, 202, 203, 204).

 

Despite suggestions that CPP is associated with subsequent development of PCOS, available data are inconsistent. Prospective longitudinal studies are needed to adequately address this concern (205, 206).

 

Who to Treat?

 

For patients less than 7 years of age with a confirmed diagnosis of CPP, the benefit of GnRHa treatment is generally unequivocal. However, the value of GnRHa treatment may be unclear for the peripubertal child (typically a girl) with onset of puberty between 7-9 years of age especially when treatment is unlikely to improve the predicted adult height (PAH) (207). Some girls and their families are comfortable with early pubertal onset and early menarche. In contrast, some girls and their families are distraught when even contemplating early puberty and premature menarche. Consistent evidence-based data regarding negative psychosocial consequences in children with CPP are lacking (208). Further, it may be challenging to justify the medical benefits of GnRHa therapy for early puberty due to the accompanying burdens of increased physician office visits and financial impact. Shared decision-making involving the patient, parents, and medical staff is indispensable to address the benefits and risks of GnRHa in the individual patient (209) . 

 

Goals of Treatment

 

Goals of GnRHa treatment include prevention of pubertal progression and height preservation (210). Growth velocity can significantly decline in some children during GnRHa treatment particularly in those with a markedly advanced bone age (211). The use of other height augmenting medications including recombinant human growth hormone (GH) (212, 213, 214, 215), stanozolol (216, 217), and oxandrolone (218) have been explored but none are recommended for sole use or as an adjunct to GnRHa therapy (219, 220). 

 

Increasing adult height must be judged considering the financial and psychological burdens of this intensive treatment regimen (221). Several recent studies have recommended treatment beyond a bone age of 12 years, however more rigorous studies are needed before such treatment is endorsed (222, 223).

 

Another goal of CPP treatment is to mitigate psychosocial distress and prevent adverse mental health outcomes. One epidemiological study of over 7000 women showed that adolescents with early age of menarche had higher rates of depression and antisocial behavior, which persisted into adulthood (224). Adverse psychosocial experiences reported in girls with early age at menarche include increased likelihood of teenage pregnancy and childbearing, sexual and physical assault, and reduced likelihood of high school graduation (225). However, studies thus far do not show that GnRHa therapy can mitigate these effects. One small study of 36 girls with CPP treated with GnRHas evaluated behavioral health diagnosis and health-related quality of life and found no abnormalities in psychological functioning (226). In a small study of 15 girls with CPP treated with GnRHa and 15 age-matched controls, comprehensive test batteries revealed similar scores in cognitive performance, behavioral, and psychosocial problems (227). A review of 15 studies evaluating the psychosocial impact of CPP showed an increased psychosocial and health-related quality of life burdens with CPP compared with controls (228). The same study showed qualitative data demonstrating emotional lability in patients with CPP and that physical differences associated with sexual precocity could increase feelings of shame and embarrassment which further increase isolation and social withdrawal (228). Again, larger studies are needed to better establish if and how GnRHas influences the psychosocial issues associated with CPP.

 

Monitoring of Treatment

 

Treatment efficacy can be monitored by repeat clinical exams assessing pubertal progression, ultrasensitive LH, FSH and sex hormone concentrations (estradiol in girls, testosterone in boys), rate of progression of bone maturation, estimates of PAH and change in PAH, and patient satisfaction. No uniform consensus exists regarding the optimal strategy for monitoring treatment efficacy in children with CPP. Progression of breast or testicular development may indicate poor adherence, treatment failure, or incorrect diagnosis (188).

 

Random basal LH concentrations to confirm treatment efficacy may be unhelpful because random LH levels often fail to revert to a prepubertal range even when the HPG axis is fully suppressed (229, 230). Therefore, random LH concentrations cannot be used to indicate treatment failure. To confirm gonadotropin suppression, a GnRH stimulation test with short-acting GnRH or, alternatively, a single LH sample 30–120 min after long-acting GnRH analog administration may be performed (231, 232) and different protocols exist regarding the specific timing and number of LH and FSH measurements (233). Some clinicians prefer to utilize clinical indices particularly in areas where hormone determinations are costly.

 

During treatment, breast tissue usually becomes softer with variable changes in size. The rate of bone maturation typically slows with adequate treatment resulting in a decline in BA/CA or a change in BA divided by time. Recent data show that the decline in BA/CA is non-linear and that larger declines are seen in the first 18 months of treatment (222). Thereafter, a slower rate of decrease suggests maintenance of suppression rather than treatment failure.

 

Height velocity is typically rapid prior to treatment and decreases on treatment. The height deceleration is most apparent during the first 18 months of treatment, similar to the deceleration in skeletal maturation. Subsequently, a prepubertal growth rate is often evident (222). Ideally, the rate of bone maturation decelerates resulting in a net gain in height potential. Therefore, calculating PAH during treatment helps assess efficacy. It is also important to understand that mid-parental height (MPH) influences height outcome. GnRHa treatment for CPP may restore genetic potential but rarely causes PAH to surpass genetic potential. Therefore, treatment efficacy by PAH assessment is always in comparison to MPH.

 

Discontinuation of Therapy

 

The decision to discontinue GnRHa treatment needs to be tailored to meet the patient’s specific needs. Factors influencing the decision-making process include synchronizing pubertal progression with peers, patient readiness for resumption of puberty, recent linear growth velocity, bone age X-ray results, and adult height prediction (234). Specific considerations for the developmentally delayed child may be reviewed with the caregivers (137, 234, 235). Pubertal manifestations generally reappear within months of discontinuation of GnRHa treatment; the mean time to menarche is approximately 16 months (217, 218). Several studies have reported that ovulatory function and menstrual cycles are normal once they resume (137, 236).

 

Eripheral Precocious Puberty or Gonadotropin-Independent Precocious Puberty

 

Peripheral precocious puberty (PPP) is due to either excessive endogenous gonadal or adrenal sex steroid secretion (estrogens or androgens) or from exogenous exposure to sex steroids. Ectopic gonadotropin secretion typically from a germ-cell tumor often located in the CNS can also lead to PPP. PPP may be appropriate for the child's sex (isosexual) or inappropriate, with virilization of females and feminization of males (heterosexual). In most instances, pubertal development is incomplete, and fertility is not attained. Etiologies of PPP include:

 

MCCUNE-ALBRIGHT SYNDROME

 

McCune-Albright syndrome (MAS) is an uncommon disorder characterized by the triad of gonadotropin-independent precocious puberty, irregular café-au-lait skin pigmentation and fibrous dysplasia of bone (237, 238).  It has been recognized more recently that MAS may exist as a “form fruste” with only one or two features (239). MAS affects both boys and girls. Importantly, precocious puberty is not observed in all affected individuals and tends to be more common among girls.

 

MAS is due to a somatic cell (post-zygotic) variant arising early during embryogenesis in the GNAS1 gene which is located at chromosome 20q13.3. This gene encodes the Gsα protein coupled to the G-protein membrane receptors for glycoprotein hormones. Vertical transmission has not been reported suggesting that germline variants are embryonic lethal. Variability in post-zygotic expression of the deleterious variant results in a mosaic pattern of tissue expression and inconsistent clinical manifestations between affected individuals (237).

 

Two missense variants, Arg201His and Arg201Cys, are the most frequently identified variants. These variants lead to loss of the α-subunit’s intrinsic GTPase activity resulting in inappropriate cyclic AMP production and constitutive receptor activation. The net result is autonomous ligand-independent signaling by LH, FSH, TSH, GHRH, and ACTH receptors leading to the associated hyperfunctioning endocrinopathies(237).

 

The café-au-lait lesions are generally large with irregular “coast of Maine” borders and typically do not cross the midline. The café-au-lait lesions result from increased tyrosinase gene expression and melanin production in affected melanocytes (240).

 

Bone manifestations are characterized by dysplastic lesions with abnormal bone turnover and inadequate mineralization. These lesions can be associated with pain, malformations, fractures, or nerve compression. The somatic cell gain-of-function variants alter the differentiation of multi-potent skeletal stem cells resulting in the replacement of normal bone and marrow with immature woven bone and fibrotic stroma. The dysplastic tissue is characterized by abundant osteoclast-like cells. Although the somatic Gsα skeletal variants arise during embryogenesis, bone development appears to be normal in utero.

 

Bony lesions become apparent during early childhood typically reaching the maximal burden in young adulthood. The variability in the somatic cell expression accounts for the variability in the location and extent of the fibrous dysplasia.  To date, an accurate ascertainment of risk to develop bone disease is unavailable. However, younger age and higher skeletal burden score derived from scintigraphic bone scans appear to predict longitudinal progression of bone disease. Importantly, evolution of bony lesions is not associated with the extent of endocrine manifestations (241).

 

Overproduction of fibroblast growth factor 23 (FGF23) by skeletal cells bearing the GNAS1 variant can lead to increased urinary phosphate excretion and decreased renal 1-α-hydroxylase activity (242). Although overt hypophosphatemic rickets is uncommon due to compensatory mechanisms, affected individuals often manifest increased serum FGF23 levels and renal phosphate wasting (243).

 

In the gonads, these variants induce ligand independent activation of gonadotropin receptors resulting in subsequent autonomous ovarian estrogen and testicular testosterone secretion in affected prepubertal girls and boys, respectively.

 

Girls may develop recurrent estrogen-secreting cysts accompanied by breast development and linear growth acceleration. Spontaneous resolution of a cyst decreases the estrogen concentration resulting in withdrawal vaginal bleeding. The sequence of pubertal development may be atypical with vaginal bleeding preceding breast development. Hence, MAS should be considered in females with recurrent ovarian cysts and vaginal withdrawal bleeding. Ovarian torsion rarely occurs. Bloodwork may reveal elevated estradiol concentrations with suppressed gonadotropin concentrations. Pelvic ultrasound typically shows one or more ovarian cysts and uterine enlargement. Nevertheless, serum estradiol concentrations and pelvic ultrasound results may be unremarkable following spontaneous involution of an ovarian cyst. Estrogen exposure may lead to accelerated skeletal maturation with adverse consequences on final adult height. In some instances, a secondary gonadotropin-dependent precocious puberty develops. In adult women, the persistent autonomous ovarian activity can lead to abnormal uterine bleeding, menometrorrhagia, which may be so severe as to require blood transfusion. Spontaneous pregnancies can occur, but relative infertility is common (244).

 

Among boys, autonomous GNAS1 activation in the testes leads to Leydig and Sertoli cell hyperplasia which can be associated with macro-orchidism. Scrotal ultrasound may show focal masses, diffuse heterogeneity, and microlithiasis. Differing from typical pubertal progression, testicular volume in boys with MAS does not accurately indicate pubertal status. Substantial autonomous testosterone production is uncommon. Approximately 15% of boys manifest clinical signs of excessive androgen secretion (239). Leydig cell hyperplasia, the most common histologic finding of the testes, carries a low risk of malignant transformation. Thus, conservative management with periodic scrotal ultrasound imaging is appropriate for follow-up of testicular masses detected in boys with MAS (245).

 

Other features associated with MAS include thyrotoxicosis, growth hormone excess (gigantism or acromegaly), and Cushing syndrome. Hypercortisolism is uncommon, typically occurs during the first year of life, and is associated with higher mortality attributed to secondary infections (246). Specific laboratory evaluation and treatment for associated endocrine features should be obtained. Genetic variants can be found in other nonendocrine organs (liver, intestines, and heart) resulting in cholestasis and/or hepatitis, intestinal polyps, and cardiac arrhythmias, respectively (247, 248). Since GNAS1 variants are considered to be weak oncogenes, the risk for malignant transformation is slightly higher than for the general population (239). In addition, women with MAS have an increased risk for breast cancer attributed to earlier estrogen exposure (249).

 

The diagnosis of MAS is usually based on the characteristic clinical features. Due to GNAS1 variant mosaicism, only 20-30% of peripheral blood lymphocytes are positive for the variant using traditional PCR-based testing. However, variant detection is greater than 80% in the affected tissues (250). Newer circulating cell free DNA testing offers a potential methodology to assess for MAS variants (251) . Importantly, negative testing, especially of peripheral blood lymphocytes, does not exclude the diagnosis of MAS.

 

Therapeutic goals focus on treating specific clinical manifestations. For manifestations related to puberty, current medications either inhibit sex steroid biosynthesis or block their actions at the level of end organs. Minimal evidence-based data are available because of the low prevalence of MAS.  Ketoconazole, an anti-fungal medication, has been used because it inhibits the steroidogenic cytochrome P450 enzymes decreasing adrenal and gonadal steroidogenesis (252). However, ketoconazole may interfere with cortisol synthesis; patients need to be monitored for possible adrenal insufficiency and may benefit from use of stress dose hydrocortisone treatment. Rarely hepatic toxicity can occur. 

 

Aromatase inhibitors prevent conversion of androgens to estrogens. Initial reports for testolactone, fadrozole, and anastrozole were disheartening because no enduring beneficial effects on skeletal growth and bone maturation were observed. Letrozole has been used and showed sustained beneficial effects on skeletal maturation and predicted final height in one small series (253).

 

Selective estrogen receptor modulators such as tamoxifen and fulvestrant have been used. Tamoxifen has both agonist and antagonist activity at the estrogen receptor. Despite reports regarding the efficacy of tamoxifen to reduce vaginal bleeding accompanied by positive effects on bone, this medication has been reported to increase risk of endometrial disease in adult women (254). In view of potential risks for endometrial cancer, tamoxifen should be used with great caution in women with MAS (255).

 

Fulvestrant is a pure estrogen receptor blocker administered by intramuscular injections at monthly intervals. In one small series, vaginal bleeding was reduced with complete cessation of vaginal bleeding in only 8/25 girls. The rate of skeletal maturation decreased without any significant change in linear growth velocity or predicted adult height. Fulvestrant was reported to be well tolerated; additional studies are needed to supplement these initial findings (256).

 

In the past, surgery cystectomy or oophorectomy had been performed in girls with MAS (257). Since cyst recurrence is common, cystectomy should be avoided if possible. Women with MAS have the potential for fertility and spontaneous pregnancy; hence, oophorectomy should be avoided (258).

 

For boys with MAS associated precocious puberty, therapeutic interventions include androgen receptor blockers, aromatase inhibitors, and ketoconazole to interfere with testosterone synthesis (258). Combination therapy with bicalutamide and anastrozole was successfully utilized in one boy with PPP due to MAS (259). Bicalutamide is a potent nonsteroidal antiandrogen that binds to and inhibits the androgen receptor and increases the receptor’s degradation. Surgical intervention should only be considered for rapidly enlarging palpable testicular masses due to the risk of malignancy (245).

 

PREMATURE MENARCHE AND OVARIAN CYSTS  

 

Functioning ovarian follicular cysts can secrete estradiol resulting in isolated premature vaginal bleeding or peripheral precocious puberty (260, 261). Additional signs of puberty may be absent in girls with isolated premature menarche. Although some girls may present with slight breast development followed by vaginal bleeding. The bleeding typically lasts only a few days and is usually attributed to spontaneous resolution/regression of an estrogen-secreting ovarian cyst. By the time a pelvic ultrasound can be obtained, the cyst has resolved, and the ultrasound shows no abnormalities. Isolated premature menarche may be limited to a single episode or may be recurrent. In most instances, linear growth velocity, onset of cyclic menstrual cycles, and final adult height are unaltered.

 

Differential diagnosis includes sexual abuse, vaginal foreign body, vaginal infections, MAS, or primary hypothyroidism(262). Due to the intermittent nature of these cysts, conservative medical management is usually appropriate (263). Large cysts may predispose to ovarian torsion (264, 265, 266, 267). Patients with ovarian torsion usually present with short duration of pain and systemic symptoms such as vomiting. Given the low frequency of malignancy in such an ovarian, detorsion with or without cystectomy is generally preferred (268). Gonadectomy should be avoided to preserve fertility. Rarely, rhabdomyosarcoma or sclerosing stromal tumors can present with vaginal bleeding.

 

OVARIAN TUMORS  

 

Estrogen-secreting ovarian tumors are a rare cause of peripheral precocious puberty. Specific types of tumors include granulosa cell, gonadal stromal cell, ovarian sex cord stromal, and theca cell tumors.

 

Juvenile granulosa cell tumors (JGCT) are the most common ovarian tumors. Typically, these tumors present with rapidly progressive isosexual precocity (269). Most JGCT are large enough to be palpated during an examination and are typically limited to the ovary at the time of diagnosis. Circulating estradiol concentrations may be extremely elevated with suppressed gonadotropin concentrations. Circulating tumor markers including α-fetoprotein (AFP), lactate dehydrogenase (LDH), β-human chorionic gonadotropin (β-hCG), cancer antigen 125 (CA-125), and inhibin B can be identified. Genetic somatic variants have been identified in juvenile granulosa cell tumors. Over 60% of JGCT carry in frame duplications in the AKT1 gene; this gene codes for a kinase involved in ovarian mitogenic signaling (270). Other identified variants include KMT2C-truncating and the ribonuclease III domain of DICER1 variants.  In contrast to adult granulosa cell tumors of the ovary, variants in the FOXL2 gene are generally not found in JGCT. Ollier and Maffucci syndromes, rare disorders associated with benign cartilaginous enchondroma, have been associated with JGCT (271). Surgical excision with peritoneal cytology for staging is the primary treatment.

 

Rarely, other tumors including gonadoblastoma, lipid tumors, cystadenomas, and ovarian carcinomas can secrete sex steroids. Finding elevated serum inhibin and AMH concentrations suggest that the tumor cells are derived from granulosa or Sertoli cells.

 

Sex cord tumors with annular tubules can occur in patients with Peutz-Jeghers Syndrome. Peutz-Jeghers Syndrome is an autosomal dominant disorder associated with mucocutaneous pigmentation, gastrointestinal polyposis, and genetic variants in the STK11 gene located at chromosome 19p13.3. (272) The gonadal tumors can be multi-focal, bilateral, and can differentiate into granulosa cell or large cell calcifying Sertoli cell tumors with the potential to secrete estrogen. Thus, girls may present with precocious puberty whereas boys may present with gynecomastia.

 

Sertoli-Leydig cell tumors are rare ovarian tumors often associated with somatic or germline DICER1 variants (273). Most are unilateral, but bilateral tumors have been described.  These tumors contain testicular structures, Sertoli and Leydig cells, and can rarely secrete androgens. Hence, girls can virilize with pubic hair development (274, 275). Girls known to carry germline DICER1 variants should undergo regular pelvic ultrasounds to screen for ovarian tumors (276).

 

LEYDIG CELL TUMORS  

 

Leydig cell tumors are a subtype of testicular stromal tumors that arise from testosterone producing Leydig cells. In prepubertal boys, presenting features include penile enlargement, acne, development of pubic and axillary hair, and accelerated linear growth velocity. Examination of the testes typically show asymmetric testicular volume due to a unilateral testicular tumor. Leydig cell tumors are usually benign. Bloodwork shows elevated circulating testosterone concentrations and suppressed gonadotropin concentrations. Ultrasound is useful to assess testicular volume and morphology.

 

Treatment involves surgical removal of the tumor. When possible, testis-sparing enucleation is preferred to radical orchiectomy to preserve testicular function and fertility. The surgical approach is dictated by the intraoperative assessment of tumor size, location, and the amount of remaining normal testicular parenchyma (277, 278).

 

HUMAN CHORIONIC GONADOTROPIN SECRETING GERM CELL TUMORS  

 

During early gestation, primordial germ cells migrate from the hindgut to the gonads. In some instances, the germ cells can migrate to locations outside of the gonad, fail to undergo apoptosis, proliferate in these atypical locations, and ultimately become hCG-secreting germ cell tumors (279). Common locations for germ cell tumors include the CNS, lung, or liver (280). Boys and men with Klinefelter syndrome are at higher risk to develop extra-gonadal GCTs particularly in the mediastinum (281). In addition, hepatoblastoma secreting hCG and α-fetoprotein can also present with precocious puberty (282). Due to the similarity between hCG and LH which have identical α-subunits and related β-subunits, tumor-derived hCG stimulates testicular LH receptors resulting in testosterone secretion. In the prepubertal boy, the aberrant hormone exposure can result in precocious puberty (27, 28). Testicular volume may not increase since seminiferous tubule growth does not occur in the absence of FSH stimulation. Bloodwork shows elevated hCG and testosterone concentrations with suppressed/variable LH and FSH concentrations.

 

Prepubertal girls generally do not develop isosexual precocious puberty with hCG-secreting germ cell tumors because in the absence of FSH, the granulosa cells do not express aromatase and are unable to synthesize estradiol.

 

GERM CELL TUMORS

 

Chromosomal aneuploidy or genetic variants can interfere with gonadal development resulting in dysgenetic gonads. In this situation, the appropriate microenvironment for normal germ cell maturation is absent, thereby disrupting the normal maturational progression for germ cells. This situation may result in the development of gonadal germ cell tumors. Precursor lesions of germ cell tumors include germ cell neoplasia in situ (GCNIS, formerly termed carcinoma in situ – CIS) and gonadoblastoma (283). Subsequently, dysgerminoma, seminoma, or non-seminoma may develop. Usually, such germ cell tumors do not secrete significant amounts of sex steroids.

 

FAMILIAL MALE LIMITED PRECOCIOUS PUBERTY (FMPP)

 

Familial male-limited precocious puberty (also known as testotoxicosis) is due to an autosomal dominant activating germline variant in the LH/choriogonadotropin receptor (LHCGR) gene located at chromosome 2p21. The LH/CG receptor is a G-protein coupled receptor (284). The variant is associated with autonomous ligand-independent receptor signaling leading to Leydig cell hyperplasia and premature testosterone secretion in prepubertal boys. Pathogenic missense variants associated with FMPP tend to congregate in an apparent hot spot located in the 6th transmembrane segment and in the 3rd intracellular loop (285).

 

Affected males typically present between two to six years of age with penile enlargement, linear growth acceleration, advanced skeletal maturation, acne, and pubarche. The testes are usually symmetrically enlarged due to the Leydig cell hyperplasia, but the size is disproportionately smaller compared to the testosterone levels (286, 287). A large portion of the testicular volume is formed by Sertoli cells which are not stimulated in this condition (286, 287). Circulating testosterone concentrations are elevated with suppressed gonadotropin concentrations. Although adult height is generally compromised, fertility has been reported (288). Longitudinal follow-up with testicular self-examination and scrotal ultrasound is recommended because malignant testicular germ cell tumors have been described in a few individuals (289).

 

Therapeutic goals include decreasing autonomous testicular testosterone secretion and slowing epiphyseal maturation. To date, several medications including ketoconazole, spironolactone, bicalutamide, and aromatase inhibitors have been used with varying efficacy (286). To date, the most effective therapy is combination treatment with an anti-androgen and an aromatase inhibitor (290, 291). If secondary GnRH-dependent precocious puberty develops, GnRH agonist therapy can be added to the therapeutic regimen. Abiraterone, a selective CYP17A1 inhibitor, was utilized in a young boy with bilateral Leydig cell tumors and resistance to the usual combination regimen of an anti-androgen and aromatase inhibitor. He required glucocorticoid replacement therapy and monitoring for possible excessive mineralocorticoid action because of abiraterone treatment-associated iatrogenic 17-hydroxylase/17,20-lyase deficiency (292).

 

Although inherited as an autosomal dominant disorder, girls do not develop precocious puberty (293). Since only the LHCGR gene is affected, the presumably minimally increased theca cell androgens cannot be aromatized to estradiol in the absence of FSH stimulation. Importantly, asymptomatic women can transmit the affected allele to their sons.   

 

PRIMARY HYPOTHYROIDISM

 

Children with profound chronic primary hypothyroidism may present with precocious puberty. Van Wyk and Grumbach described this association in 1960 (294). Clinical features in girls include early breast development, vaginal bleeding, and galactorrhea. Boys present with testicular enlargement. Pubic and axillary hair are absent. Typical features associated with hypothyroidism such as short stature, impaired linear growth, puffy face, dry skin, constipation, and delayed skeletal maturation (despite pubertal changes) are usually evident. Pituitary imaging shows an enlarged pituitary gland. Abdominal ultrasound may show ovarian enlargement with or without ovarian cysts. Labs show mild elevation in FSH levels but LH levels usually remain prepubertal.

 

Levothyroxine therapy induces regression of pubertal symptoms, stops vaginal bleeding, and decreases pituitary volume. However, final height may often be compromised due to accelerated skeletal maturation upon initiation of thyroxine treatment. One potential mechanism for the precocious puberty is cross-reactivity of TSH at the ovarian FSH receptor. TSH and FSH share a common α-subunit with hormone specificity due to the differing β-subunits (295). This mechanism was tested using recombinant human TSH in an in vitro bioassay which, at high concentrations, was able to stimulate human FSH receptors (296, 297, 298, 299).

 

VIRILIIZING CONGENITAL ADRENAL HYPERPLASIAS

 

The virilizing congenital adrenal hyperplasias (CAHs) are autosomal recessive disorders associated with impaired adrenal steroidogenesis due to genetic variants in steroidogenic enzyme genes. The most common is 21-hydroxylase deficiency due to variants in the 21-hydroxylase gene (CYP21A2) located at chromosome 6p21.33. Clinically, congenital adrenal hyperplasias reflect a phenotypic spectrum ranging from presentation in neonatal period with classic salt-losing CAH to presentation during infancy/todder age with classic simple virilizing CAH to later presentations with non-classic CAH. Milder or non-classic forms have been described for 11-β-hydroxylase deficiency and 3β-hydroxysteroid dehydrogenase deficiency (300).

 

Children with non-classic CAH typically present with premature pubarche characterized by pubic/axillary hair development, acne, accelerated linear growth velocity, and advanced skeletal maturation. Girls may have clitoromegaly whereas boys may have phallic enlargement with prepubertal testicular volume. Adult women with non-classic CAH usually present with irregular menses, hirsutism, and infertility.

 

The diagnostic test for 21-hydroxylase deficiency is an elevated 17-hydroxyprogesterone (17-OHP) concentration. Early morning basal 17-OHP values have been suggested as an effective screening test with reports of 100% sensitivity and 99% specificity with a threshold value of 200 ng/dl (6 nmol/L) to diagnose NCAH in children who present with premature pubarche (301). If the diagnosis is highly suspected despite relatively normal 17-OHP concentrations, an ACTH stimulation test may be indicated to exclude the diagnosis of 21-hydroxylase deficiency. For an ACTH stimulation test, following collection of a basal blood sample, 0.25 mg synthetic ACTH (Cortrosyn) is administered by intravenous or intramuscular routes; a second blood sample is collected at 30 and/or 60 minutes. Physician preference governs the timing of the ACTH-stimulated 17-OHP concentration. In the future, 21-deoxycortisol and 11-oxyandrogens may be increasingly utilized in the diagnosis and management of 21-hydroxylase deficiency (302, 303). The reader is referred to more extensive discussion of the virilizing CAH (304, 305).

 

ADRENOCORTICAL TUMORS

 

Androgen-secreting adrenocortical tumors are extremely rare causes of PPP accounting for less than 1% of all childhood malignancies. Most tumors occur in children younger than 4 years of age with a second smaller peak in adolescents. Pediatric adrenocortical tumors are categorized as adenomas or carcinomas based on histological features. However, histopathologic differentiation may be challenging, and biologic behavior of the tumor may help with this categorization (306).

 

Pediatric adrenocortical carcinoma is more common in girls than boys and has a bimodal pattern with peaks under age 5 and over 10 years of age (307). Adrenocortical tumors are associated with several genetic syndromes such as Li-Fraumeni syndrome and Beckwith-Wiedemann syndrome (BWS). Li-Fraumeni Syndrome is an autosomal dominant familial cancer syndrome associated with germline p53 gene variants. The p53 gene (or TP53 gene) is a tumor suppressor gene located at chromosome 17p13.1, and codes for the protein p53. Malignancies associated with Li-Fraumeni syndrome include adrenocortical carcinomas, breast cancer, brain tumors, and sarcoma. The incidence of adrenocortical tumors is 10-15 times higher in southern Brazil; this has been attributed to the higher prevalence of the R337H variant of the TP53 gene (308).

 

Beckwith-Wiedemann syndrome is characterized by macroglossia, macrosomia, organomegaly, neonatal hypoglycemia due to hyperinsulinism, and abdominal wall defects. This disorder is associated with uniparental disomy in the 11p15 chromosomal region leading to IGF2 growth factor overexpression. Although only 1% of children with Beckwith-Wiedemann Syndrome will develop adrenocortical carcinomas, these adrenal tumors account for approximately 20% of the neoplasms in children with this disorder (309). Other disorders associated with adrenal tumors include Multiple Endocrine Neoplasia Syndrome Type 1 (MEN1) and Carney complex (310).

 

The next section reviews variants of puberty associated with early pubertal changes and are important differentials to consider in the evaluation of CPP.

.

PREMATURE THELARCHE

 

Premature thelarche is the premature development of glandular breast development. The breast development may be unilateral or bilateral. Typically, premature thelarche develops in otherwise healthy girls between 12-24 months of age and is self-limited. No other pubertal changes are evident; linear growth velocity is normal and pubic/axillary hair are absent. On physical examination, the areolae and vaginal mucosa are prepubertal. The diagnosis can usually be made on a clinical basis without bloodwork or bone age X-rays (311). Pelvic ultrasound showed increased prevalence of ovarian microcysts in girls with premature thelarche compared to age-matched controls; no correlation between ovarian cysts, gonadotropin concentrations, and estradiol concentrations has been found (312). Longitudinal follow-up is appropriate to confirm the diagnosis and assess for the unlikely possibility of progression to CPP.

 

PREMATURE ADRENARCHE

 

Pubarche refers to the appearance of pubic/axillary hair, increased apocrine odor, and acne due to the onset of adrenarche. Adrenarche refers to the pubertal maturation of the adrenal zona reticularis. Adrenarche which normally occurs in children between 6-8 years of age and is characterized by increased secretion of the adrenal androgen precursors DHEA, DHEAS, and androstenedione.

 

Premature adrenarche is characterized by premature pubarche, which is defined as the development of pubic or axillary hair before 8 years in girls or 9 years in boys. There is no breast development in females and no testicular enlargement in males. Bone age is usually not advanced. Premature adrenarche is a diagnosis of exclusion. Thus, exclusion of other disorders such as CAH, androgen-secreting tumors, exogenous androgen exposures, and other rare genetic disorders such as apparent cortisone reductase and PAPS synthase 2 (PAPSS2) deficiencies is essential (313).

 

Children with premature adrenarche and early androgen excess may be at a higher risk to develop the metabolic syndrome. Waist circumference (WC), waist/hip ratio, and total and truncal fat mass increase are detected in premature adrenarche. Increases in systolic and diastolic blood pressure (BP), total cholesterol (TC), very low-density lipoprotein (VLDL), TC/high density lipoprotein (HDL), low density lipoprotein (LDL)/HDL ratio, and atherogenic index (AI) have been reported. Increased insulin concentrations starting from prepubertal ages may occur suggesting that premature adrenarche may be one of the first symptom of insulin resistance (IR) in childhood (314, 315). T2DM may occur in a subset of these cases. Ovarian hyperandrogenism, hirsutism, ovulatory dysfunction, and polycystic ovaries may be more frequent in girls with premature adrenarche during post pubertal ages than normal population. Although, early retrospective data in a homogenous population suggests an association between premature adrenarche and adolescent hyperandrogenism (316), more recent longitudinal data suggests that premature adrenarche was not associated with adolescent ovarian dysfunction and was only associated with lower SHBG concentrations (317).

 

EXPOSURE TO EXOGENOUS SEX STERIODS  

 

Feminization, including gynecomastia in males, has been attributed to excess estrogen exposure from creams, ointments, and sprays. Other possible sources of estrogen exposure include contamination of food with hormones, phytoestrogens (e.g., in soy), and over-the-counter remedies such as lavender oil and tea tree oil (318, 319, 320, 321). Similarly, virilization of young children has been described following inadvertent exposure to androgen-containing creams/gels (322).

 

Endocrine-Disrupting Chemicals 

 

Various endocrine-disrupting chemicals (EDCs) are found in the environment  (323, 324, 325, 326).  Most EDCs have chemical structures similar to those of endogenous sex steroids. These chemicals can disrupt steroid hormone receptor binding and hormone metabolism altering hormone concentrations or changing hormone synthesis/degradation (327). EDCs can act beyond steroid hormone receptors by affecting transcriptional modulators and direct effects on genes. Some EDCs have mixed activities, and most EDCs include several different chemicals. The patient’s age and duration of exposure modulate the consequences of EDC exposure. In addition, EDCs can be classified as persistent (long-lasting) or non-persistent (short half-lives). Environmental EDC exposures can be transgenerational such that future generations could be affected (328). Mechanisms for EDC exposures include ingestion, topical use, inhalation, and transfer across the placenta (329).

 

The consequences of mixed “cocktail” EDC exposures on pubertal development are indeterminate. A systematic review with a stringent meta-analysis found no consistent association between xenobiotic EDCs and pubertal timing apart from an insinuation that, in girls, postnatal exposure to phthalates could be associated with earlier thelarche and later pubarche, consistent with their anti-androgenic properties. Methodological heterogeneity, limited number of studies, and variability in statistical analyses constrained the conclusions of this systematic review. Hence, future longitudinal epidemiologic studies to clarify the specific EDCs, age at exposure, and duration of exposure will be valuable (327, 330).

 

 

DELAYED PUBERTY

 

Gonadarche associated with the reactivation of the GnRH pulse generator, is signified by breast development in girls and testicular enlargement in boys. Delayed puberty is defined as absence or delayed onset of gonadarche at a chronologic age >2 standard deviations later than the population mean. In girls, delayed puberty is defined as absence of breast development by age 13 years or lack of menarche by age 15 years (331) or 3 years from onset of thelarche (332). In boys, delayed puberty is defined as the lack of testicular enlargement to a volume >= 4 ml by age 14 years (333). Delayed puberty is more common in boys than in girls.

 

Four main categories of delayed puberty have been described (See Table 4).

  • transient hypogonadotropic hypogonadism associated with delayed maturation of the HPG axis also known as constitutional delay of growth and puberty (CDGP)
  • hypergonadotropic hypogonadism characterized by primary gonadal dysfunction with consequent elevated LH and FSH concentrations.
  • hypogonadotropic hypogonadism with low LH and FSH concentrations due to congenital (CHH) or acquired causes
  • functional hypogonadotropic hypogonadism (FHH), as seen in chronic health disorders such as cystic fibrosis, renal failure, inflammatory bowel disease, restrictive eating disorders etc.

 

Table 4. Etiologies of Delayed Puberty (458)

Condition

Etiology

Constitutional Delay of Growth and Puberty

Genetic basis has infrequently been described in HS6ST1, FTO, IGSF10, EAP1 genes

Hypergonadotropic Hypogonadism

Congenital:

-Klinefelter’s syndrome

-Turner syndrome

-Gonadal dysgenesis

-Anorchia

-Primary ovarian insufficiency

-Testicular regression syndrome

-Genetic causes: FMR1, STAG3, NR0B1, NR5A1, FOXL2, WT1 and others

-Galactosemia

Acquired:

-Infectious (mumps)

-Autoimmune (polyglandular syndromes)

-Surgery (torsion, trauma)

-Chemotherapy (alkylating agents)

-Radiation

-Gonadal tumor

Hypogonadotropic Hypogonadism

Congenital:

-Isolated HH: Over 50 genes have been identified; notable are ANOS1, FGFR1, FGF8, PROK2, CHD7, KISS1, KISS1R, GNRH, GNRHR and others

-Prader Willi

-CHARGE syndrome

-Noonan

-Bardet-Biedl

- Panhypopituitarism associated with genetic variants in PROP1, HESX1, LHX, LHB, FSHB and others.

Acquired:

-Central nervous system tumors (e.g., craniopharyngiomas, germinomas), cysts,

-Cranial surgeries,

-Cranial radiation therapy greater than

 30 Gy

-Other inflammatory, autoimmune (hypophysitis), and infiltrative (Langerhans cell histiocytosis) diseases of the pituitary gland

Functional Hypogonadotropic Hypogonadism

-intense physical stress (competitive gymnastics, ballerina syndrome)

-emotional stress (elevated glucocorticoids)

-caloric deficit (anorexia nervosa)

-chronic systemic illness (celiac, inflammatory bowel disease, CF, renal disease)

-endocrinopathies (hypothyroidism, excess glucocorticoids, hyperprolactinemia)

-medication adverse effects

-pituitary iron deposits in chronic transfusion dependent children

 

Transient Hypogonadotropic Hypogonadism Associated with Delayed Maturation of the HPG Axis/ Constitutional Delay of Growth and Puberty (CDGP)

 

Transient hypogonadotropic hypogonadism also known as constitutional delay in growth and puberty (CDGP) is the most common etiology of delayed puberty occurring in 70% of boys and 32% of girls with delayed puberty (334). In both sexes, CDGP is self-limited and is considered to represent a variant of normal pubertal timing. CDGP has a strong genetic component, with a positive family history of delayed puberty reported in 50% to 75% of cases (335).

 

Distinguishing between CDGP and congenital hypogonadotropic hypogonadism (CHH) may be challenging because these conditions share clinical features, hormone levels, and radiological findings. Inhibin B and LH levels tend to be lower in boys with CDGP, but the overlap in values precludes the use of these hormones to distinguish between CDGP and CHH.

 

For boys, 3-4 months of steroid priming with testosterone followed by 3-4 months of observation is commonly used to discriminate CHH from CDGP (336). It has been suggested that this sex steroid exposure stimulates resumption of the HPG axis activity leading to secondary sex characteristics typical of male puberty (337). Individuals who show no pubertal progression during the observation period should be evaluated for CHH or another disorder affecting the HPG axis. Estradiol priming has been used similarly in girls to distinguish CHH from CDGP (338). Due to the differences in the long-term outcomes, accurate diagnosis is essential, with CDGP being largely a diagnosis of exclusion (339).

 

CDGP occurs more commonly in family members of individuals with CHH compared to the general population (340). Individuals with CDGP appear to have higher prevalence of pathogenic variants compared to unaffected family members or controls (341). Some genetic variants have been detected in both individuals with CDGP and CHH; these genes include HS6ST1, PROKR2, TAC3, TAC3R, and IL17RD (342). Genetic variants associated primarily with CDGP include IGFS10, EAP1, and FTO (338).

 

Hypergonadotropic Hypogonadism

 

Pubertal delay associated with hypergonadotropic hypogonadism is usually associated with disorders affecting gonadal function, specifically gonadal steroidogenesis. With the onset of gonadarche and increased GnRH and gonadotropin secretion, inadequate gonadal steroid secretion and lack of negative feedback leads to increasing gonadotropin secretion. These conditions may be present at birth or acquired.

 

TURNER SYNDROME  

 

Turner Syndrome refers to deletions or structural rearrangements of the X chromosome. The

reported incidence is around 1 in 2500 liveborn female births (343). The initial in utero process of ovarian differentiation proceeds normally with migration of the primordial germ cells into the developing ovary during the fourth week of gestation. By 18 weeks of gestation, premature degeneration of ovarian follicles has begun. The ovarian follicles are typically replaced by connective tissue resulting in the characteristic streak gonad. This accelerated follicular atresia usually leads to premature ovarian insufficiency. Girls with Turner syndrome have gonadal dysgenesis or “streak gonads” in 85% of cases at birth. However, because adrenal androgen secretion is not impaired, the onset of pubarche usually occurs at a normal time. Typical clinical features of girls with Turner syndrome include short stature, short/webbed neck, shield shaped chest with the appearance of widely spaced nipples, cubitus valgus, and Madelung deformity of the forearm and wrist, shortened fourth metacarpals/metatarsals, horseshoe kidneys, coarctation of the aorta, increased risk for autoimmune conditions, and aberrant development of the lymphatic system. Many girls with Turner syndrome may remain undiagnosed until later in childhood or adolescence when they present with short stature and/or delayed puberty. With increased utilization of noninvasive prenatal screening (NIPS), many girls with Turner Syndrome are detected prenatally. The reader is referred to other publications for more extensive discussion regarding the features and approach to multidisciplinary health care management (344, 345, 346).

 

KLINEFELTER SYNDROME

 

Klinefelter Syndrome is a chromosomal aneuploidy characterized by 47, XXY karyotype and premature testicular insufficiency. Increased NIPS utilization has led to detection of many boys in utero and is estimated to occur in 1 in 667 males based on prenatal cytogenetic analysis (347). However, many men remain underdiagnosed, with less than 10% of patients being diagnosed prior to puberty. Men with Klinefelter syndrome typically present with tall stature, incomplete puberty, or gynecomastia. Generally, the onset of puberty is not delayed. Klinefelter syndrome is associated with small firm testes, Sertoli cell dysgenesis, impaired spermatogenesis, and variable degrees of testosterone deficiency (348). Learning disabilities, language and visuospatial processing defects, and neuropsychiatric conditions such as attention-deficit/hyperactivity disorder and depression are common (349). If a tumor is found in the anterior mediastinum, a karyotype should be performed to evaluate for Klinefelter syndrome because of its association with mediastinal germinoma (350). Despite normal BMI, the body fat percentage, and the ratio between android fat percentage and gynoid fat percentage are significantly higher than normal. They may also have an impaired bone metabolism starting during childhood and adolescence. Systematic studies are needed to evaluate whether testosterone replacement therapy during puberty will improve these parameters (351). The reader is referred to other publications for more extensive discussion regarding the features and approach to multidisciplinary health care management (352, 353, 354, 355, 356).

 

DIFFERENCES OF SEX DEVELOPMENT  

 

Differences of sex development (DSDs) are a group of conditions where external genital development is atypical. These disorders are associated with chromosomal anomalies, genetic variants, and environmental influences (357). Gonadal function is impaired in some types of DSDs resulting in primary gonadal failure and hypergonadotropic hypogonadism. Detailed review of DSDs is beyond the scope of this chapter. The interested reader is referred to other Endotext chapters for more extensive review of DSDs.

 

GENETIC CAUSES OF PREMATURE OVARIAN INSUFFICIENCY (POI)

 

POI can present with primary or secondary amenorrhea.  Fragile X-associated premature ovarian insufficiency is among a family of disorders caused by the expansion of a CGG trinucleotide repeat sequence located in the 5′ untranslated region (UTR) of the fragile X messenger ribonucleoprotein 1 (FMR1) gene on the X chromosome. One etiology is premutation of the FMR1 gene associated with 55-200 CGG repeats without abnormal methylation of the neighboring CpG island and promoter, responsible for both fragile X associated premature ovarian insufficiency in females and fragile X associated tremor ataxia syndrome in males and females where patients may present with mild to moderate intellectual disability, intentional tremor and cerebellar ataxia, peripheral neuropathy, Parkinsonism, and urinary and bowel incontinence.

 

The X chromosome carries many genes that govern follicular maturation and overall ovarian function, and numerical and structural changes in this chromosome, as in Turner syndrome or triple X syndrome, are associated with POI.

 

Multiple genes are involved in ovarian differentiation, oocyte development, and, ultimately, folliculogenesis and variants in these genes may be associated with premature ovarian insufficiency (358, 359). The clinical phenotype ranges from delayed puberty to secondary amenorrhea (360). 

 

GALACTOSEMIA

 

Galactosemia is a rare cause of delayed puberty. Classic galactosemia is a rare inborn error of galactose metabolism due to a defect in the gene encoding the galactose-1-phospate uridyltransferase enzyme (GALT). The prevalence is approximately 1/30,000–60,000 (361). Early manifestations include lactose intolerance, jaundice, failure to thrive, lethargy, hepatocellular damage, renal tubular disease, and cataracts. A galactose-free diet can reverse the neonatal symptoms. However, some long-term complications such as developmental delay, intellectual disability, epilepsy, osteoporosis, and premature ovarian insufficiency may still develop. In females, hypergonadotropic hypogonadism resulting in delayed puberty, primary or secondary amenorrhea, and infertility may occur (362, 363). Available data from patients with classic galactosemia suggest that the primary ovarian insufficiency is due to dysregulation of pathways essential for folliculogenesis culminating in premature ovarian insufficiency (364). Several previous cohort studies in males showed delayed puberty and below-target final height (365, 366, 367), however a recent study with 47 males showed that puberty and fertility were normal and in contrast to earlier reports, AMH, testosterone and Inhibin B levels were normal (361).

 

Hypogonadotropic Hypogonadism

 

Pubertal delay associated with hypogonadotropic hypogonadism is usually associated with disorders affecting the neurons that secrete GnRH or the pituitary gonadotrophs that secrete the FSH and LH. These conditions may be present at birth or acquired as described below.

 

CONGENITAL HYPOGONADOTROPIC HYPOGONADISM

 

The initiation and maintenance of reproductive capacity in humans depends on pulsatile GnRH secretion. Congenital hypogonadotropic hypogonadism (CHH) results from the absence of the normal pulsatile GnRH secretion or deficient pituitary gonadotropin secretion leading to delayed puberty and infertility. The number of genetic loci associated with CHH continues to expand (Table 4). CHH may be associated with variants in genes involved in the development or migration of GnRH neurons as well as genes involved in the secretion or action of GnRH (368). Autosomal recessive, autosomal dominant, X-linked, and oligogenic inheritance have been described (369, 370). Additional genetic influences include epigenetic factors (371). Clinical heterogeneity has been described between and within families (372).

 

Given the developmental origins of GnRH neurons in the olfactory placode, CHH can be associated with anosmia or hyposmia. The association of CHH and anosmia is known as Kallmann syndrome. Classic Kallmann syndrome is associated with variants in the ANOS1 gene which is mapped to the X chromosome. Other features of Kallmann syndrome due to ANOS1 variants include unilateral renal agenesis, sensorineural hearing loss, dental agenesis, synkinesia (alternating mirror movements), and cleft lip/palate (373).

 

In syndromic CHH, associated clinical features may help identify the possible gene(s). For example, clinical features associated with FGFR1 variants include anosmia/hyposmia, cleft lip/cleft palate, dental agenesis, and skeletal anomalies. CHH can also occur in the CHARGE syndrome, which is characterized by coloboma, congenital heart disease, choanal atresia, genital anomalies, ear anomalies, and development delay. CHH can occur with impaired pituitary development associated with PROP1, HESX1, or LHX variants. CHH is also associated with variants in the GnRH, GnRHR, LHB, and FSHB genes (See Table 5). Although some genetic loci are common to both CDGP and CHH, the genetic architectures of these two conditions are largely distinct (374).

 

Table 5. Genes Associated with Delayed Puberty (338, 482, 483)

Gene

(Reference/s)

Protein encoded

Genetic locus

Associated features/syndromes

                                         SYNDROMIC CAUSES

FGFR1/FGF8

 

(484, 485)

Fibroblast Growth Factor Receptor 1/fibroblast growth factor 8

8p11.23

Hartsfield syndrome

LEPR/LEP

(486, 487, 488)

Leptin receptor and Leptin

1p31.3

Severe obesity syndromes

PCSK1

(489)

Prohormone convertase 1 gene

5q15

Obesity, ACTH deficiency, diabetes

DMXL2

(490)

DmX-like protein 2

15q21

Polyendocrinopathy, Polyneuropathy syndrome

RNF216/

OTUD4

(491)

Ring finger protein 216/ OTU domain-containing protein 4

4q31.21

Gordon Holmes

PNPLA6

(492, 493)

Patatin-like phospholipase domain-containing protein 6

19p13.2

Gordon Holmes, Oliver McFarlane,

Lawrence Moon, Boucher-Neuhauser syndrome

SOX10

(494)

Sex determining region Y-Box transcription factor 10

22q13.1  

Wardenburg syndrome

SOX2

(495)

Sex determining region Y-Box transcription factor 2

3q26.33

Optic nerve hypoplasia, CNS abnormalities

SOX3

(496)

Sex determining region Y-Box transcription factor 3

Xq27.1

Intellectual disability, craniofacial abnormalities, multiple pituitary hormone deficiencies

IGSF1

(497, 498)

Immunoglobulin superfamily member 1

Xq26

Associated with X-linked central hypothyroidism, macro-orchidism

HESX1

(499)

HESX homeobox 1

3p14.3

Hypopituitarism, septo-optic dysplasia

CHD7

(500, 501, 502)

Chromodomain helicase DNA binding protein 7

8q12.2

CHARGE syndrome

POLR3A/

POLR3B

(503, 504, 505)

 

RNA polymerase III

12q23.3

Hypomyelination, hypodontia

NROB1

(DAX-1)

 

(506, 507)

Nuclear Receptor Subfamily 0 Group B Member 1/ dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1

Xp21

Adrenal hypoplasia

REV3L/

PLXND1

(508)

 

Catalytic subunit of DNA polymerase zeta

6q21

Möbius syndrome

PWS

(509, 510)

 

15q11.2

Prader-Willi syndrome

BBS1, BBS2, ARL6, BBS4, BBS5, MKKS, BBS7, TTC8, BBS9, BBS10,

TRIM32, BBS12

(511, 512)

Encoded protein may play a role in eye, limb, cardiac and reproductive system development

11q13.2, 20p12, 16q21, 15q22.3‐23, 14q32.1

(multiple loci)

Bardet-Biedl syndrome

PHF6

(513)

Plant homeodomain (PHD)-like finger protein 6 

Xp26.2

Borjeson‐Forssman‐Lehmann syndrome

SMCHD1

 

(514)

Structural maintenance of chromosomes flexible hinge domain containing 1

18p11.32

Bosma arhinia microphthalmia syndrome

TBC1D20/

RAB18

(459, 515)

TBC1 Domain Family Member 20, GTPase activator proteins of Rab-like small GTPases

 

20p13

Warburg micro syndrome

HDAC8

(516)

 

Histone deacetylase 8

Xq13.1

Cornelia de Lange syndrome

                                          NON-SYNDROMIC CAUSES

FGF17

(517)

Fibroblast Growth Factor 17

8p21.3

 

ANOS1 (KAL1)

(518, 519)

Kallmann syndrome protein, which is now known as Anosmin 1

Xp22.31 

involved in fibroblast growth factor (FGF) signaling

GNRHR/

GNRH1

(520, 521, 522)

 

Gonadotropin-releasing hormone receptor/ gonadotropin-releasing hormone 1

4q13.2

 

KISS1R/

KISS1

(54, 523)

Kisspeptin-1 receptor/ kisspeptin-1

19p13.3

 

 KLB

(524)

Klotho Beta

4p14

Metabolic defects

TAC3/TACR3

(342, 525)

Tachykinin 3, Tachykinin 3 receptor

Encodes neurokinin b

4q24

 

IL17RD

(517)

 

Interleukin 17 Receptor D

3p14.3

 

DUSP6

(517)

 

Dual specificity phosphatase 6

12q22–q23

 

SEMA3A/

SEMA3E/

SEMA7A

(526)

Semaphorin 3A

7q21.11

 

SPRY4

(517)

Sprouty homolog 2

5q31.3

 

FLRT3

(517)

Fibronectin leucine rich transmembrane protein 3

20p11

 

PROKR2/

PROK2

(527, 528)

Prokineticin-2 and Prokineticin receptor 2

3p13

 

WDR11

(529)

 

WD repeat domain 11

10p26.12

 

 

CCDC141

(530)

Coiled-Coil Domain Containing 141

2q31.2

 

FEZF1

(531)

FEZ family zinc finger 1

7q31.32

 

LHB

(532)

Luteinizing hormone

19q13.33

 

FSHB

(533, 534)

Follicle-stimulating hormone

11p14.1

 

AXL

(458)

 

AXL receptor tyrosine kinase

19q13.2

 

EAP1

(535)

Enhanced at puberty 1

14q24

Trans-activates the GnRH promoter

LGR4

(536)

Receptor for R-spondins which, once activated, potentiates the canonical Wnt signaling pathway

11p14.1

 

TUBB3

(483)

Microtubule protein β-III-tubulin

16q24.3

Congenital fibrosis of the extraocular muscles

WDR11, PROP1, PROK2, PROKR2

(529)

Bromodomain and WD repeat-containing protein 2, Homeobox protein 

prophet of PIT-1, prokinectin 2

10q26.12,

5q35.3,

3p13

Combined pituitary hormone deficiency

FTO

(482, 537)

Fat mass and obesity-associated protein

16q12.2

Mice lacking FTO had significantly delay in pubertal onset

 

ACQUIRED HYPOGONADOTROPIC HYPOGONADISM

 

Several conditions are associated with primary gonadal insufficiency. These conditions include auto-immune disorders, trauma, neoplasia, vascular events, and infection. Autoimmune disorders can be associated with premature ovarian and testicular insufficiency. Biallelic mutations in the autoimmune regulator (AIRE) gene are associated with autoimmune polyendocrine syndrome type 1 which is also known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Associated features include mucocutaneous candidiasis, hypoparathyroidism, and adrenal insufficiency (375). The detection of autoantibodies directed against tissue-specific antigens suggests an autoimmune diagnosis.   

 

Antineoplastic chemotherapy with alkylating agents, as well as localized ionizing radiation, may permanently damage germ cells leading to infertility. In males, Sertoli cells are more susceptible to such toxicity than Leydig cells such that testosterone production may remain intact despite Sertoli and germ cell injury. Mumps orchitis should be considered, especially in unvaccinated males. Decreased blood flow to the gonads from surgical injury (e.g., orchiopexy in boys), torsion, or trauma can lead to ischemia and atrophy, with resultant primary testicular insufficiency.

 

The presence of otherwise normal male external genitalia associated with nonpalpable gonads indicates that the testes were present and functioning at least early in gestation. The “vanishing testes syndrome” also known as testicular regression is associated with atrophy or regression of testicular tissue initially formed during early embryonic development. Potential etiologies of the testicular regression include in utero vascular disruption or testicular torsion. Pathogenic variants of the DEAH-box RNA helicase DHX37 (DHX37) gene have been identified in boys with testicular regression and in association with gonadal dysgenesis (376).

 

In addition to autoimmune etiologies, premature ovarian insufficiency can be associated with ovarian/pelvic tumors, chemotherapy, especially alkylating agents, and radiation therapy. The location of the pelvic tumor and treatments influence the ovarian reserve and risk for premature ovarian insufficiency. Low or declining serum AMH levels provide an indirect measure of ovarian reserve. However, due to much variability and lack of diagnostic thresholds, measuring AMH values does not accurately predict ovarian insufficiency in cancer survivors (377).

 

Acquired hypogonadotropic hypogonadism (HH) can be due to central nervous system tumors such as craniopharyngiomas and germ cell tumors. Such tumors can disrupt the hypothalamic-pituitary stalk or can impact pituitary function producing decreased gonadotropin production. Hyperprolactinemia due to prolactin-secreting adenomas can cause acquired HH (378). Other central nervous system disorders associated with acquired HH include hypophysitis, histiocytosis, and hemochromatosis. Intracranial surgeries and/or cranial radiation therapy greater than 30 Gy are known risk factors for HH. Moderate to severe trauma to the brain is associated with injuries to the hypothalamus, stalk (infundibulum), or pituitary gland itself; the consequences of traumatic brain injury may not manifest for many years. Chronic steroid treatment can be associated with acquired HH in boys with Duchenne muscular dystrophy (379, 380). Inflammatory and infiltrative diseases of the pituitary gland are other rare causes of acquired HH.

 

FUNCTIONAL HYPOGONADOTROPIC HYPOGONADISM

 

Functional HH is the hypothalamic response to intense physical or emotional stress, caloric deficit, or chronic systemic illness (381). In this situation, the otherwise normal HPG axis fails to function due to the concomitant stress. Puberty can be delayed or stalled until the underlying condition has been adequately addressed. The finding of hypercortisolemia in women with functional HH associated with restrictive eating disorders highlights the relevance of HPA axis function in FHH (382). Importantly, functional HH can have long lasting adverse consequences on bone health (383).

 

The hypothalamus receives numerous inputs regarding body energy status and subsequently modulates reproductive status based on this information. Hence, nutritional status and energy output influence HPG axis activity in part via leptin signaling which regulates the sensitivity of the pituitary to GnRH (384). Energy deficits may occur due to weight loss, excessive energy expenditure (rigorous physical activity, renal disease, cystic fibrosis, congenital heart disease), decreased caloric intake or malabsorption (disordered eating behaviors, bowel disorders such as celiac, Crohn’s, and ulcerative colitis) are associated with delayed or stalled puberty and  functional hypothalamic amenorrhea (385, 386, 387). Elevated circulating levels of cytokines (as seen in some acute or inflammatory conditions) may also inhibit the HPG axis. Elevated prolactin levels, due to prolactinoma or severe primary hypothyroidism may inhibit gonadotropin release.

 

Some boys with obesity have low gonadotropin and testosterone levels and manifest delayed puberty (388). It is important to recognize that certain medications such as antipsychotics (typical and atypical), certain antidepressants, and opioids can alter menses (364). 

 

Treatment of Delayed Puberty

 

A variety of therapeutic regimens for pubertal induction have been described for both boys and girls. However, large, randomized trials providing evidence-based data regarding the optimum regimen are lacking (389). Sex steroid replacement therapy remains a mainstay of treatment. The type and route of administration of the sex steroids is dependent on patient preference, insurance coverage, and health care provider practices. Importantly, the specific treatment regimen depends on the underlying etiology of the pubertal delay. Future novel therapies could include kisspeptin and neurokinin B analogs (390).

 

BOYS

 

Pulsatile GnRH therapy is the most physiological method and can induce adult secondary sex characteristics, achieving normal adult testosterone concentrations, and spermatogenesis (370) in boys with HH. However, the inconvenience of wearing a mini-pump and conflicting outcome data limits its usefulness. Other approaches include hCG, FSH, hMG, and/or GnRH treatments. Despite much heterogeneity, a systematic study reported that treatment with hCG and FSH induced greater increase in testicular volume and rate of spermatogenesis compared to hCG alone (391)370). Importantly, available limited data suggest that testosterone administration prior to gonadotropin treatments does not interfere with the beneficial effects on testicular growth and spermatogenesis. Based on the physiologic roles of LH and FSH, pubertal induction should begin with FSH to promote testicular maturation followed by combined FSH and hCG treatment. The subsequent hCG treatment will promote testicular testosterone secretion leading to virilization, growth spurt, and psychosocial development.

 

Still, at the present time, testosterone is the most established treatment for pubertal induction in boys with delayed puberty. Traditionally, IM testosterone esters, primarily testosterone enanthate or testosterone cypionate, have been used. A subcutaneous testosterone enanthate auto-injector has recently been approved, but this approach requires more weekly injections and is more expensive. However, no evidence-based guidelines exist for testosterone-induced pubertal initiation. Potential adverse consequences of testosterone therapy include erythrocytosis, premature epiphyseal closure especially with excessive doses which may result in aggressive behavior, mood swings, and priapism.

 

Other testosterone formulations include testosterone gels, pills, and pellets. Limitations of testosterone gels include difficulties in accurately titrating low doses, potential testosterone exposure to household members, and the cost. Oral methyltestosterone and its 17α-derivatives have been associated with hepatic dysfunction and should be avoided. Oral testosterone undecanoate was approved by the FDA in 2019 to treat hypogonadal adult men. However, due to its short half-life, multiple daily doses are necessary, and no data are available regarding use for puberty induction. Testosterone pellets require surgical placement every 3-4 months, are expensive, and often spontaneously extrude (392).

 

For the younger adolescent boy with a strong family history of CDGP, reassurance and continued clinical monitoring may be adequate. However, discerning CDGP from CHH is essential because the treatment, genetics, and psychosocial implications differ. Hence, low dose testosterone for 3-4 months followed by a similar period of observation may be helpful to distinguish CDGP from CHH. Individuals with CHH will show persistently low gonadotropin and sex steroid hormone levels after the 3–4-month period of observation whereas individuals with CDGP will usually show spontaneous pubertal progression. Curiously, testosterone exposure apparently activates GnRH production and secretion leading to “reversal” with onset of HPG axis activity in some boys with CHH. This reversal is associated with specific genetic variants and may be transient (393, 394).

 

GIRLS

 

Timely induction of pubertal development is fundamental. Two major goals of estrogen therapy are mimicking typical pubertal progression with breast development and promoting adequate uterine growth (395). Although pulsatile GnRH treatment can be used, this approach has no advantage over estrogen for pubertal induction in girls. Though all therapeutic approaches utilize estrogens, details regarding specific formulations and methods of administration vary. Transdermal estradiol is preferred for replacement therapy because this approach avoids the first pass through the liver and the potential for adverse effects on clotting factors.

 

Typically, low transdermal estradiol doses are used for the initial phase of pubertal induction. Transdermal estradiol doses of 3-7 mcg/day can be achieved by cutting matrix patches (0.014-0.025 mg/24 h) into quarters or eighths. Subsequently, the dose can be increased approximately every six months until adult replacement dosage is achieved taking about 24-36 months to do so. High initial estrogen doses should be avoided due to increased likelihood for atypical breast development characterized by prominent nipples with little supporting breast tissue. High estrogen doses should also be avoided as premature epiphyseal fusion could impair additional linear growth.

 

Oral micronized 17β-estradiol can be used for those with severe skin irritation or aversion to the use of a patch. Oral preparations containing conjugated equine estrogens or ethinyl estradiol should be avoided for both pubertal induction and maintenance therapy. Most combined oral contraceptives contain ethinyl estradiol at doses higher than appropriate for induction of puberty. Approximately 18-24 months after initiation of unopposed estrogen therapy, progestogens can be added to induce withdrawal bleeding and to reduce the risk for endometrial hyperplasia. Progestogens can be introduced earlier if breakthrough vaginal bleeding occurs. Pelvic ultrasounds before and during pubertal induction can be planned to assess uterine size and shape as well as to evaluate endometrial thickness to ascertain optimal timing to introduce progestins. Progestins vary in potency and can be administered by transdermal, oral, or uterine routes. Although increased potency may have beneficial effects on withdrawal bleeding, greater progestogenic side effects may develop.

 

No evidence-based data exist, and no single regimen has been demonstrated to be superior. Pubertal induction therapy should be individualized based on clinical response and other auxologic parameters.

 

Oral contraceptive pills may be used for convenience but should be limited to after completion of pubertal development. Since some girls may experience sporadic ovulation, contraception should be utilized by those at risk of undesired pregnancies.

 

EVALUATION OF A CHILD WITH A VARIATION IN PUBERTAL DEVELOPMENT

 

The diagnostic tools are comparable for the evaluation of either precocious or delayed puberty. Detailed medical history and physical examination provide the preliminary information to guide the differential diagnosis for a child with a variation in pubertal development (165, 396) (see Figures 5-8). Laboratory, imaging, and genetic studies are subsequently utilized to ascertain the specific diagnosis. The tools for evaluation of a child with a variation in pubertal development are described below. The tools are comparable, but the interpretation of test results differs for precocious and delayed puberty.

 

Figure 5. Algorithm to evaluate a girl presenting with precocious puberty. *follow clinical progression every 3-6 months. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CAH: Congenital Adrenal Hyperplasia; ACTH: Adrenocorticotrophic Hormone; GnRH: Gonadotropin Releasing Hormone; DHEAS: Dehydroepiandrosterone Sulfate; 17OHP: 17-hydroxy progesterone; NF-1: Neurofibromatosis-1.

Figure 6. Algorithm to evaluate a boy presenting with precocious puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; MRI: Magnetic Resonance Imaging.

Figure 7. Algorithm to evaluate a girl presenting with delayed puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CDGP: Constitutional Delay in Growth and Puberty; CHH: Congenital Hypogonadotropic Hypogonadism; MRI: Magnetic Resonance Imaging.

Figure 8. Algorithm to evaluate a boy presenting with delayed puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CDGP: Constitutional Delay in Growth and Puberty; CHH: Congenital Hypogonadotropic Hypogonadism; MRI: Magnetic Resonance Imaging.

 

History and Physical Examination

 

The medical history focuses on the timing and sequence of the pubertal changes in the patient as well as parents, grandparents, and siblings. Review of past medical history and medications (including chemotherapy and nutritional supplements) is essential. Inquiry regarding exposures (tea tree/lavender oils, sex steroids, radiation) may help to identify potential environmental factors (162). Obtaining birth history, length and weight, history of SGA (397), prematurity, or CNS insult at birth or later provide relevant information (162).

 

A history of gelastic seizures may point to a hypothalamic hamartoma. Inquiry regarding use of transdermal testosterone by a family member may identify the cause of premature virilization. Pubertal delay associated with micropenis, anosmia, cryptorchidism, deafness, choanal atresia, hearing loss, and/or digital abnormalities suggests congenital hypogonadotropic hypogonadism (CHH). A family history of anosmia, subfertility, and deafness should be sought for those with pubertal delay. Multiple syndromes are associated with CHH (see Table 3); suggestive features include absent/reduced sense of smell, choanal atresia, hearing loss, morbid obesity, visual impairment. Family history of precocious or delayed puberty in close relatives may be discovered (398). Behavioral difficulties or learning disabilities may be associated with specific syndromes such as Turner or Klinefelter syndromes.

 

A complete physical examination including height, weight, arm span, and sitting height is essential. Review of the child’s growth curves provides valuable information regarding changes in linear growth and weight gain. Acceleration in linear growth and upward crossing of centiles may be seen in precocious puberty. A gradual downward crossing of centiles may be noted in constitutional delay in growth and puberty (CDGP) as linear growth slows compared to peers who are entering puberty (399). Pubic hair development (pubarche) may also be delayed in CDGP as opposed to CHH where adrenarche occurs at the normal age for population (372).

 

Physical exam includes ascertainment of the sexual maturity rating for breast, pubic hair, and testicular volume based on the scoring system derived by Tanner and colleagues (Figure 1). Due to challenges in discriminating lipomastia from true glandular breast development, palpation of the breasts is important. Firm glandular tissue under the areolae is indicative of thelarche. Accurate measurement of testicular volume using an orchidometer is essential (see Figure 2). A testicular volume of ≤ 1.1 mL has a reported sensitivity and specificity of 100% and 91%, respectively, for CHH (400).

 

The physical examination should assess for midline defects, dysmorphic features, visual field abnormalities, and features characteristic for specific syndrome. For example, short stature, cubitus valgus, low hair line, widely spaced nipples, and delayed puberty suggest Turner Syndrome. The physical examination needs to include palpation of the thyroid gland, skin examination for acne or café-au-lait macules (which would suggest neurofibromatosis or McCune-Albright syndrome) and a visual field exam. Melanocytic macules typical of Peutz-Jeghers syndrome could point to the presence of a sex cord tumor causing gonadotropin independent (peripheral) sexual precocity.

 

Laboratory Evaluation

 

Laboratory evaluation assists the diagnostic process to identify the etiology of “off-time puberty.” Circulating gonadotropin and sex steroid concentrations reflect HPG axis status (187, 236). Most current gonadotropin assays are sandwich assays specific to the β-subunit. Ultrasensitive FSH and LH assays should be used when available. For LH, samples should preferably be obtained in the morning. The lower limit of detection for most ultrasensitive immunochemiluminescent assays (ICMA) is ≤0.1 mIU/mL (230, 401, 402).

 

When the clinical concern is precocious puberty, LH concentrations greater than 0.3-0.5 mIU/mL suggest central precocious puberty (CPP) with higher cut-points increasing the sensitivity and specificity of the LH determination (403). Elevated basal LH levels show high sensitivity and specificity for boys when high quality immunochemiluminometric assays (ICMA) is used (404). Different cut-points need to be used to interpret LH concentrations in girls under two years of age because LH concentrations may be elevated at this age leading to misdiagnosis of CPP followed by inappropriate treatment during this phase of development (405). For the child with physical signs of premature puberty, LH concentrations in the prepubertal range are consistent with either peripheral precocity or a benign pubertal variant such as premature thelarche. Typically, LH and FSH concentrations are suppressed in children with peripheral precocious puberty (406).

 

In the evaluation for delayed puberty, low gonadotropin concentrations suggest a central etiology such as CDGP or hypogonadotropic hypogonadism while elevated gonadotropin concentrations suggest primary gonadal insufficiency. Random gonadotropin concentrations may provide only limited information because gonadotropin secretion is pulsatile. Distinguishing hypogonadotropic hypogonadism from CDGP is often challenging because LH, FSH and sex hormone reference intervals vary widely even in healthy adolescents (407). Similarly, due to significant overlap in hormone reference intervals, GnRH agonist and human chorionic gonadotropin (hCG) stimulated gonadotropin (408) and sex steroid concentrations fail to distinguish youth with CHH from those with CDGP (407, 409).

 

Due to the small structural differences between steroid molecules, immunoassays are confounded by cross-reactivity issues. Assay issues are amplified in children because commercial immunoassays for estradiol and testosterone are usually designed to measure hormone concentrations within the normal adult reference interval. Hence, most estradiol immunoassays have low sensitivity and specificity to quantify the low concentrations (< 30 pg/ml) typically found in prepubertal children and individuals with hypogonadism. Similar issues occur with testosterone immunoassays. Hence, steroid hormone concentrations should be measured by liquid chromatographic separation followed by mass spectrometry (LC-MS/MS). Serum testosterone is best measured using LC-MS/MS technology to limit cross-reactivity and increase sensitivity and specificity especially when low hormone concentrations might be anticipated. LC-MS/MS is also the optimal technique to measure circulating concentrations of other steroids including 17-hydroxyprogesterone, DHEA, androstenedione, and the 11-oxy androgens. It offers greater sensitivity and specificity and allows simultaneous measurement of multiple hormone concentrations (410).

 

Sex steroids such as estradiol and testosterone circulate bound to sex hormone binding globulin (SHBG). Tissue availability of the free hormone, presumed to be the active moiety, is regulated by SHBG. Direct free testosterone concentrations should be avoided because direct immunoassays have poor reproducibility and reliability. When free testosterone concentrations need to be determined, equilibrium dialysis should be performed despite known potential limitations including increased expense, reliance on total testosterone accuracy, temperature control, and sample dilution (411).

 

Another confounding factor is biotin (vitamin B7) which is an over-the counter supplement by itself or as an addition to many preparations used to strengthen nails and hair. Biotin interferes with the technical aspects of immunoassays and can lead to either falsely elevated or falsely low result when streptavidin binding is utilized in the assay detection system. When immunoassay results seem incongruous, use of biotin-containing products should be queried. Biotin does not interfere with LC-MS/MS assays (412).

 

GnRH or GnRH AGONIST STIMULATION TEST

 

Historically, the established gold standard to diagnosis CPP was the LH and FSH response to a standard bolus of native GnRH. With decreased availability of native GnRH, most stimulation tests are now performed with the GnRH agonist (GnRHa) leuprolide acetate, a synthetic nonapeptide with much greater potency. The timing and peak values of FSH and LH levels differ between GnRH and leuprolide acetate. Following native GnRH administration, LH levels peak after 20–40 minutes, followed by a decline. With leuprolide acetate, peak LH occurs between 0.5 - 4 hours followed by sustained LH elevation.

 

The optimal cutoff value of peak stimulated LH for identifying children with CPP is unclear due to assay variability. For most LH assays, a value of 3.3 to 5 mIU/mL defines the upper limit of normal for stimulated LH values in prepubertal children. Stimulated LH concentrations above this range suggest CPP (232). Children with progressive CPP tend to have a high stimulated LH:FSH ratios compared with those with non- or intermittently progressive precocious puberty. Measuring the appropriate sex steroid 24 hours following GnRHa administration can help confirm a CPP diagnosis (413). However, obtaining this second sample may burden the family because of the need for a second venipuncture, expense of another hormone determination, and missed school and work.

 

As noted above regarding basal LH levels, care must be taken in interpreting the results of GnRH stimulation test in females under the age of two years, as both basal and stimulated LH levels can be elevated as part of the normal hormonal changes associated with mini-puberty (405).

 

To assess GnRH production by the hypothalamus, kisspeptin-stimulated LH response has been proposed to identify individuals with GnRH deficiency and thus CHH. Kisspeptin stimulates GnRH secretion, thus promoting LH, and to a lesser extent FSH, secretion.

 

One study found that maximal LH rise after kisspeptin administration was more accurate for diagnosis of men with GnRH deficiency than GnRH stimulation testing (414). A similar study in adolescents with pubertal delay (3 females and 13 males), peak LH post kisspeptin stimulation was demonstrated to be superior to GnRH stimulation testing for predicting capacity to progress through puberty (noting that the LH cut off values were different and an ideal cutoff value still needs to be determined) (346). Further research is required to better define the parameters of using kisspeptin stimulation in clinical practice (404).

 

In children with precocious pubarche, measurement of adrenal steroids may be necessary to help distinguish between peripheral precocity and benign premature adrenarche. Children with premature adrenarche can have mild elevation in adrenal hormones (415). Since premature adrenarche is a diagnosis of exclusion, further investigation for congenital adrenal hyperplasia and virilizing adrenal tumors may be indicated. In children, an early-morning 17-hydroxyprogesterone (17-OHP) value >200 ng/dL (6 nmol/L) has a high sensitivity and specificity for non-classic congenital adrenal hyperplasia secondary to 21-hydroxylase deficiency. An adrenocorticotropic hormone (ACTH) stimulation test is needed to confirm the diagnosis (313, 416). An ACTH stimulation test involves administration of 0.25 mg synthetic ACTH (1-24) or 15 mcg/kg for children up to 2 years of age with blood samples obtained at baseline and either 30 or 60 minutes after synthetic ACTH administration. Although 21-hydroxylase deficiency is the most common virilizing form of CAH, 17-hydroxypregnenolone, DHEA, and 11-deoxycortisol determinations may be necessary to assess for 3β-hydroxysteroid dehydrogenase or 11β-hydroxylase deficiencies.

 

Boys with hypogonadotropic hypogonadism tend to have lower inhibin B values compared to boys with CDGP. However, a validated cut-point for inhibin B concentrations remains to be established (417, 418, 419). FSH stimulated inhibin B concentrations < 116 pmol/L have been demonstrated in a study of adolescents with delayed puberty to have more accurate diagnostic discrimination and a promising test for prediction of onset of puberty (414).

 

Human chorionic gonadotropin concentrations can be measured in males to evaluate for the possibility of an hCG-secreting tumor leading to peripheral precocity (280). A thyroid-stimulating hormone (TSH) concentration should be measured if chronic primary hypothyroidism is suspected as the underlying cause for the sexual precocity, known as the Van-Wyk-Grumbach syndrome (296, 298) .

 

Targeted diagnostic tests are warranted in some cases to investigate for specific causes of apparent functional hypogonadotropic hypogonadism, such as anti-transglutaminase IgA for celiac disease. Despite promising data, measurement of AMH and INSL3 in addition to testosterone, as endocrine markers to guide the differential diagnosis (418, 420), need additional studies  

 

The testosterone response to long-term hCG stimulation and peak serum FSH response to GnRH were found to be significantly different in CHH patients (421). However, there are potential long-term drawbacks to prolonged hCG therapy in males who are FSH-naïve regarding premature stimulation of Sertoli and germ cell differentiation prior to FSH exposure (338). 

 

Imaging Studies  

 

BONE AGE

 

Assessing the skeletal maturation based on a radiograph of the left hand and wrist is an important diagnostic tool in pubertal evaluation. For the commonly utilized Greulich and Pyle method, the patient’s bone age radiograph is compared with an atlas of radiographs from children of known ages (422). For the Tanner-Whitehouse 2 method, 20 different hand and wrist bones are scored. Bone age standards are largely based on hand and wrist radiographs obtained from children of European ancestry between the 1930s to the 60s (423). Despite this limitation, the bone age radiograph is a valuable indicator regarding sex steroid exposure and epiphysial (growth plate) maturation. Additional factors such as other hormones, obesity, genetics, nutritional status, various disease states, and certain medications can influence the rate of epiphyseal maturation (424) (425).

 

Bone age has been used to predict adult height using the tables of Bayley and Pinneau (426), but reliability is low with a tendency toward overestimation (427). The use of automated measurement systems with artificial intelligence has increased, mitigating previous limitations due to intra- and inter-observer variability (428, 429). Bone age readings within two standard deviations of the chronologic age are considered to be within normal limits. A delayed bone age is usually observed in patients with delayed puberty and an advanced bone age is observed with precocious puberty. One exception is patients with precocious puberty associated with hypothyroidism (Van Wyk Grumbach syndrome) where the bone age is delayed despite pubertal changes. In some instances, monitoring the predicted adult height (PAH) during the course of treatment of pubertal disorders helps to assess treatment efficacy.

 

ULTRASOUND IMAGING

 

In females, pelvic ultrasound is a rapid, non-invasive, and relatively low-cost method to ascertain the anatomy of the ovaries and uterus, ovarian volume, and uterine development. This imaging is generally readily accessible and does not require sedation, radiation, or use of contrast material. However, the quality of the device and operator experience influence the analysis.

 

During puberty, increased gonadotropin secretion promotes ovarian growth, increased estradiol secretion, and increased uterine volume (430). Girls with CPP have increased uterine size and ovarian volumes compared to prepubertal girls or those with premature thelarche. However, the overlap between prepubertal and early pubertal girls for ovarian volume and uterine size confounds interpretation of the ultrasound findings (431) (432). In a prepubertal patient with isolated vaginal bleeding, a normal pelvic ultrasound does not exclude the diagnosis of a functional ovarian cyst because the cyst may have regressed prior to imaging. Pelvic ultrasounds should be obtained in girls with primary amenorrhea who fail progesterone withdrawal to assess for Mullerian duct and renal anomalies. For patients with rapid development of secondary sex characteristics, pelvic ultrasound studies may be needed to assess for gonadal tumors.

 

The use of Doppler ultrasound to assess utero-ovarian blood flow may also provide helpful information. With increased estradiol secretion and stimulation of the estrogen receptors, vascular resistance of the uterine arteries is reduced. The pulsatility index (PI) is defined as the difference between peak systolic flow and end-diastolic flow divided by the mean flow velocity; the PI reflects impedance to blood flow distal to the sampling point. A review showed that PI is lower among pubertal girls. However, definitive cut-points for PI values have not been established. In addition, testing is operator dependent (433, 434, 435, 436).

 

Ultrasound examination of the testes, especially if asymmetric in size, should be performed in males with peripheral precocity to evaluate for the possibility of a Leydig cell tumor (437, 438). Testicular ultrasound imaging should be performed regularly to assess for testicular rest tissue in boys with congenital adrenal hyperplasia (439).

 

MR AND CT IMAGING

 

Brain MR or CT imaging is performed to define brain and pituitary anatomy. Brain and pituitary MR is helpful to assess for intracranial pathology among those with CNS symptoms. Most studies recommend a contrast-enhanced brain MRI for girls with onset of secondary sexual characteristics before six years of age because of higher rates of CNS abnormalities in these patients (137). In a 2018 meta-analysis (440), the prevalence of intracranial lesions was 3 percent among girls presenting with CPP after six years of age, compared with 25 percent among those presenting before six years. Thus, girls with pubertal onset between six and eight years of age may not need the MRI in the absence of clinical evidence of CNS pathology (441, 442, 443). MRI should be limited to high-risk individuals (younger age, neurologic symptoms) (444). Current guidelines recommend that in otherwise asymptomatic girls with CPP, a discussion occur with the parents regarding the pros and cons of brain imaging and assist in informed decision making (137, 445, 446). While contrast-enhanced brain MRIs are recommended for all boys presenting with CPP (412), one study found that these rates may be overestimated. The prevalence of intracranial lesions among boys who were healthy, did not have neurological symptoms, and were diagnosed with CPP was lower than that previously reported and none of the identified lesions necessitated treatment, suggesting the need to globally reevaluate the prevalence of pathological brain lesions among boys with CPP (447).

 

For children with delayed puberty, MR imaging of the pituitary gland and olfactory structures can assess for features of CHH such as absence of the olfactory bulbs (448, 449, 450).

 

Pelvic MRI is helpful to characterize and stage pediatric ovarian masses due to excellent soft tissue contrast. In addition, MR imaging does not involve the use of ionizing radiation and allows better assessment of the abdomen and kidneys. Disadvantages of MRI include that it is time-consuming, expensive, and may require sedation.

 

In both girls and boys, adrenal tumors can cause peripheral precocious puberty, progressive virilization, and/or markedly elevated serum adrenal androgens (e.g., DHEAS). If diagnoses such as congenital adrenal hyperplasia and exogenous androgen or testosterone exposure have been excluded, such patients should have an imaging study of the adrenal glands (451, 452, 453). CT may be preferable for evaluation, staging, surgical planning for adrenal tumors (454). Despite radiation exposure, CT can be readily performed in emergent situations.

 

Genetic Testing  

 

A karyotype can help with a diagnosis of Turner or Klinefelter syndrome (455).Newer sequencing technologies along with increased knowledge regarding genes involved in puberty has advanced the usefulness of genetic testing (456). The known genetic causes of CPP and HH have increased exponentially over the past five years. Genetic testing could therefore precede brain MRI, at least in familial CPP cases (167, 457).

 

Patients with delayed puberty associated with phenotypic features such as anosmia/hyposmia, synkinesia, or hearing loss, the probability of detecting a pathogenic variant on genetic testing for HH is increased (458) (459, 460). Consideration should be given to using genetic testing early in the diagnostic process while recognizing the limitations of genetic testing. Challenges in using genetic testing as a discriminatory test between CHH and CDGP remain, and more research is needed in this area.

 

SUMMARY

 

Pubertal development and maturation of the neuroendocrine system involve the ontogeny, activity, and interactions of the GnRH neurons. Pubertal onset is accompanied by an increase in kisspeptin and neurokinin B secretion regulating the pulsatile GnRH secretion that stimulates pulsatile pituitary LH and FSH secretion. LH and FSH stimulate gonadal sex steroid secretion promoting development of secondary sex characteristics and influencing hypothalamic-pituitary function via negative feedback inhibition.

 

Alterations of gut microbiome at different pubertal stages may present an area for future development in the prediction and prevention of precocious puberty. Use of genetic testing including targeted next generation sequencing and whole exome sequencing may have increasing utility as diagnostic tools early on in the evaluation of pubertal disorders.

 

Discovery regarding the details of normal reproductive physiology followed by identification of the genetic basis for disorders of pubertal timing established our current knowledge base for the evaluation and management of children with disorders affecting the timing of puberty. Despite the vast expansion of our knowledge, much remains to be learned about the physiology and regulation of the HPG axis from the fetus to the young adult.

 

REFERENCES

 

  1. Springfield: Mass, G. & C. Merriam Co; 1961. Webster’s third new international dictionary of the English language, unabridged.
  2. Argente J, Dunkel L, Kaiser UB, Latronico AC, Lomniczi A, Soriano-Guillen L, et al. Molecular basis of normal and pathological puberty: from basic mechanisms to clinical implications. Lancet Diabetes Endocrinol. 2023;11(3):203-16.
  3. Abreu AP. Unveiling the central regulation of pubertal development. J Clin Endocrinol Metab. 2023.
  4. Reiter EO, Fuldauer VG, Root AW. Secretion of the adrenal androgen, dehydroepiandrosterone sulfate, during normal infancy, childhood, and adolescence, in sick infants, and in children with endocrinologic abnormalities. J Pediatr. 1977;90(5):766-70.
  5. Witchel SF BA, Oberfield SE. Adrenal Androgens, Adrenarche and Adrenopause. 8th Edition ed. Robertson RP GL, Grossman A, Hammer GD, Jensen MD, Kahaly GJ, Swerdloff R, Thakker RV (eds), editor: Elsevier; 2021.
  6. Rege J, Turcu AF, Kasa-Vubu JZ, Lerario AM, Auchus GC, Auchus RJ, et al. 11-Ketotestosterone Is the Dominant Circulating Bioactive Androgen During Normal and Premature Adrenarche. J Clin Endocrinol Metab. 2018;103(12):4589-98.
  7. Counts DR, Pescovitz OH, Barnes KM, Hench KD, Chrousos GP, Sherins RJ, et al. Dissociation of adrenarche and gonadarche in precocious puberty and in isolated hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 1987;64(6):1174-8.
  8. Conley AJ, Moeller BC, Nguyen AD, Stanley SD, Plant TM, Abbott DH. Defining adrenarche in the rhesus macaque (Macaca mulatta), a non-human primate model for adrenal androgen secretion. Mol Cell Endocrinol. 2011;336(1-2):110-6.
  9. Cutler GB, Jr., Glenn M, Bush M, Hodgen GD, Graham CE, Loriaux DL. Adrenarche: a survey of rodents, domestic animals, and primates. Endocrinology. 1978;103(6):2112-8.
  10. Cumberland AL, Hirst JJ, Badoer E, Wudy SA, Greaves RF, Zacharin M, et al. The Enigma of the Adrenarche: Identifying the Early Life Mechanisms and Possible Role in Postnatal Brain Development. Int J Mol Sci. 2021;22(9).
  11. Remer T, Boye KR, Hartmann MF, Wudy SA. Urinary markers of adrenarche: reference values in healthy subjects, aged 3-18 years. J Clin Endocrinol Metab. 2005;90(4):2015-21.
  12. Marshall WA, Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child. 1969;44(235):291-303.
  13. Marshall WA, Tanner JM. Variations in the pattern of pubertal changes in boys. Arch Dis Child. 1970;45(239):13-23.
  14. Fuqua JS, Eugster EA. History of Puberty: Normal and Precocious. Horm Res Paediatr. 2022;95(6):568-78.
  15. Bruserud IS, Roelants M, Oehme NHB, Madsen A, Eide GE, Bjerknes R, et al. References for Ultrasound Staging of Breast Maturation, Tanner Breast Staging, Pubic Hair, and Menarche in Norwegian Girls. J Clin Endocrinol Metab. 2020;105(5):1599-607.
  16. Pedersen JL, Nysom K, Jorgensen M, Nielsen CT, Muller J, Keiding N, et al. Spermaturia and puberty. Arch Dis Child. 1993;69(3):384-7.
  17. Mieritz MG, Raket LL, Hagen CP, Nielsen JE, Talman ML, Petersen JH, et al. A Longitudinal Study of Growth, Sex Steroids, and IGF-1 in Boys With Physiological Gynecomastia. J Clin Endocrinol Metab. 2015;100(10):3752-9.
  18. David K DE, Freud J, Laqueur E. Crystalline male hormone from the testes (Testosterone) is more effective than androsterone derived from urine or cholesterin. Hoppe-Seyler Z Physiol Chem. 1935:233:81–2.
  19. Butenandt A HG. Umwandlung des Dehydroandrosterons in Androstendiol und Testosteron; ein Weg zur Darstellung des Testosterons aus Cholesterin. Hoppe-Seyler Z Physiol Chem. 1935:237:89–98.
  20. Ruzicka L WA. Synthetische Darstellung des Testikelhormons Testosteron (Androsten 3-on-17-ol). Helv Chim Acta. 1935:18:1264–75.
  21. Doisy EA TS, Veler CD The Crystals of the Follicular Ovarian Hormone. Proceedings of the Society for Experimental Biology and Medicine. 1930(27(5):417-419).
  22. Maccorquodale DW, Thayer SA, Doisy EA. The Crystalline Ovarian Follicular Hormone. Proceedings of the Society for Experimental Biology and Medicine. 1935;32:1182 -
  23. Smith PE. Ablation and transplantation of the hypophysis in the rat. Anat Rec. 1926;32(221):15-6.
  24. Zondek B. Uber die hormone des hypophysenvorderlappes. I. Wachstums Hormon, follikelreifungshormon (Prolan A), Luteinisierungshormon (Prolan B) Stoffwechselhormon. Klin Wochschr. 1930(9(6):245–8 ).
  25. H. L. Fevold FLH, and S. L. Leonard. THE GONAD STIMULATING AND THE LUTEINIZING HORMONES OF THE ANTERIOR LOBE OF THE HYPOPHESI. The American Journal of Physiology. 1931:97.2.291.
  26. Harris GW. The Induction of Ovulation in the Rabbit, by Electrical Stimulation of the Hypothalamo-hypophysial Mechanism. Proceedings of The Royal Society B: Biological Sciences. 1937;122:374-94.
  27. Schally AV, Arimura A, Kastin AJ, Matsuo H, Baba Y, Redding TW, et al. Gonadotropin-releasing hormone: one polypeptide regulates secretion of luteinizing and follicle-stimulating hormones. Science. 1971;173(4001):1036-8.
  28. Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E. Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. Science. 1978;202(4368):631-3.
  29. Burgus R, Butcher M, Ling N, Monahan M, Rivier J, Fellows R, et al. [Molecular structure of the hypothalamic factor (LRF) of ovine origin monitoring the secretion of pituitary gonadotropic hormone of luteinization (LH)]. C R Acad Hebd Seances Acad Sci D. 1971;273(18):1611-3.
  30. Plant TM. Recognition that sustained pituitary gonadotropin secretion requires pulsatile GnRH stimulation: a Pittsburgh Saga. F S Rep. 2023;4(2 Suppl):3-7.
  31. Plant TM, Steiner RA. The fifty years following the discovery of gonadotropin-releasing hormone. J Neuroendocrinol. 2022;34(5):e13141.
  32. Kaplan SL, Grumbach MM. The ontogenesis of human foetal hormones. II. Luteinizing hormone (LH) and follicle stimulating hormone (FSH). Acta Endocrinol (Copenh). 1976;81(4):808-29.
  33. Winter JS, Faiman C, Hobson WC, Prasad AV, Reyes FI. Pituitary-gonadal relations in infancy. I. Patterns of serum gonadotropin concentrations from birth to four years of age in man and chimpanzee. J Clin Endocrinol Metab. 1975;40(4):545-51.
  34. Casoni F, Malone SA, Belle M, Luzzati F, Collier F, Allet C, et al. Development of the neurons controlling fertility in humans: new insights from 3D imaging and transparent fetal brains. Development. 2016;143(21):3969-81.
  35. Taroc EZM, Naik AS, Lin JM, Peterson NB, Keefe DL, Jr., Genis E, et al. Gli3 Regulates Vomeronasal Neurogenesis, Olfactory Ensheathing Cell Formation, and GnRH-1 Neuronal Migration. J Neurosci. 2020;40(2):311-26.
  36. Duittoz AH, Forni PE, Giacobini P, Golan M, Mollard P, Negron AL, et al. Development of the gonadotropin-releasing hormone system. J Neuroendocrinol. 2022;34(5):e13087.
  37. Fueshko SM, Key S, Wray S. GABA inhibits migration of luteinizing hormone-releasing hormone neurons in embryonic olfactory explants. J Neurosci. 1998;18(7):2560-9.
  38. Lund C, Pulli K, Yellapragada V, Giacobini P, Lundin K, Vuoristo S, et al. Development of Gonadotropin-Releasing Hormone-Secreting Neurons from Human Pluripotent Stem Cells. Stem Cell Reports. 2016;7(2):149-57.
  39. Yellapragada V, Eskici N, Wang Y, Madhusudan S, Vaaralahti K, Tuuri T, et al. FGF8-FGFR1 signaling regulates human GnRH neuron differentiation in a time- and dose-dependent manner. Dis Model Mech. 2022;15(8).
  40. Wang Y, Madhusudan S, Cotellessa L, Kvist J, Eskici N, Yellapragada V, et al. Deciphering the Transcriptional Landscape of Human Pluripotent Stem Cell-Derived GnRH Neurons: The Role of Wnt Signaling in Patterning the Neural Fate. Stem Cells. 2022;40(12):1107-21.
  41. Seeburg PH, Adelman JP. Characterization of cDNA for precursor of human luteinizing hormone releasing hormone. Nature. 1984;311(5987):666-8.
  42. Hagen C, McNeilly AS. The gonadotrophins and their subunits in foetal pituitary glands and circulation. J Steroid Biochem. 1977;8(5):537-44.
  43. Clements JA, Reyes FI, Winter JS, Faiman C. Studies on human sexual development. III. Fetal pituitary and serum, and amniotic fluid concentrations of LH, CG, and FSH. J Clin Endocrinol Metab. 1976;42(1):9-19.
  44. Guimiot F, Chevrier L, Dreux S, Chevenne D, Caraty A, Delezoide AL, et al. Negative fetal FSH/LH regulation in late pregnancy is associated with declined kisspeptin/KISS1R expression in the tuberal hypothalamus. J Clin Endocrinol Metab. 2012;97(12):E2221-9.
  45. Kaplan SL, Grumbach MM. Pituitary and placental gonadotrophins and sex steroids in the human and sub-human primate fetus. Clin Endocrinol Metab. 1978;7(3):487-511.
  46. Althumairy D, Zhang X, Baez N, Barisas G, Roess DA, Bousfield GR, et al. Glycoprotein G-protein Coupled Receptors in Disease: Luteinizing Hormone Receptors and Follicle Stimulating Hormone Receptors. Diseases. 2020;8(3).
  47. Andersson AM, Skakkebaek NE. Serum inhibin B levels during male childhood and puberty. Mol Cell Endocrinol. 2001;180(1-2):103-7.
  48. Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, et al. Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid. Nature. 1986;321(6072):776-9.
  49. Wijayarathna R, de Kretser DM. Activins in reproductive biology and beyond. Hum Reprod Update. 2016;22(3):342-57.
  50. Abbara A, Koysombat K, Phylactou M, Eng PC, Clarke S, Comninos AN, et al. Insulin-like peptide 3 (INSL3) in congenital hypogonadotrophic hypogonadism (CHH) in boys with delayed puberty and adult men. Front Endocrinol (Lausanne). 2022;13:1076984.
  51. Crowley WF, Jr., McArthur JW. Simulation of the normal menstrual cycle in Kallman's syndrome by pulsatile administration of luteinizing hormone-releasing hormone (LHRH). J Clin Endocrinol Metab. 1980;51(1):173-5.
  52. Wildt L, Marshall G, Knobil E. Experimental induction of puberty in the infantile female rhesus monkey. Science. 1980;207(4437):1373-5.
  53. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr., Shagoury JK, et al. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614-27.
  54. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972-6.
  55. Plant TM. The neurobiological mechanism underlying hypothalamic GnRH pulse generation: the role of kisspeptin neurons in the arcuate nucleus. F1000Res. 2019;8.
  56. Herbison AE. The Gonadotropin-Releasing Hormone Pulse Generator. Endocrinology. 2018;159(11):3723-36.
  57. Cheng G, Coolen LM, Padmanabhan V, Goodman RL, Lehman MN. The kisspeptin/neurokinin B/dynorphin (KNDy) cell population of the arcuate nucleus: sex differences and effects of prenatal testosterone in sheep. Endocrinology. 2010;151(1):301-11.
  58. Nagae M, Uenoyama Y, Okamoto S, Tsuchida H, Ikegami K, Goto T, et al. Direct evidence that KNDy neurons maintain gonadotropin pulses and folliculogenesis as the GnRH pulse generator. Proc Natl Acad Sci U S A. 2021;118(5).
  59. Abreu AP, Dauber A, Macedo DB, Noel SD, Brito VN, Gill JC, et al. Central precocious puberty caused by mutations in the imprinted gene MKRN3. N Engl J Med. 2013;368(26):2467-75.
  60. Busch AS, Hagen CP, Almstrup K, Juul A. Circulating MKRN3 Levels Decline During Puberty in Healthy Boys. J Clin Endocrinol Metab. 2016;101(6):2588-93.
  61. Hagen CP, Sorensen K, Mieritz MG, Johannsen TH, Almstrup K, Juul A. Circulating MKRN3 levels decline prior to pubertal onset and through puberty: a longitudinal study of healthy girls. J Clin Endocrinol Metab. 2015;100(5):1920-6.
  62. Varimo T, Dunkel L, Vaaralahti K, Miettinen PJ, Hero M, Raivio T. Circulating makorin ring finger protein 3 levels decline in boys before the clinical onset of puberty. Eur J Endocrinol. 2016;174(6):785-90.
  63. Palumbo S, Cirillo G, Aiello F, Papparella A, Miraglia Del Giudice E, Grandone A. MKRN3 role in regulating pubertal onset: the state of art of functional studies. Front Endocrinol (Lausanne). 2022;13:991322.
  64. Li C, Lu W, Yang L, Li Z, Zhou X, Guo R, et al. MKRN3 regulates the epigenetic switch of mammalian puberty via ubiquitination of MBD3. Natl Sci Rev. 2020;7(3):671-85.
  65. Abreu AP, Toro CA, Song YB, Navarro VM, Bosch MA, Eren A, et al. MKRN3 inhibits the reproductive axis through actions in kisspeptin-expressing neurons. J Clin Invest. 2020;130(8):4486-500.
  66. Heras V, Sangiao-Alvarellos S, Manfredi-Lozano M, Sanchez-Tapia MJ, Ruiz-Pino F, Roa J, et al. Hypothalamic miR-30 regulates puberty onset via repression of the puberty-suppressing factor, Mkrn3. PLoS Biol. 2019;17(11):e3000532.
  67. Naule L, Mancini A, Pereira SA, Gassaway BM, Lydeard JR, Magnotto JC, et al. MKRN3 inhibits puberty onset via interaction with IGF2BP1 and regulation of hypothalamic plasticity. JCI Insight. 2023;8(8).
  68. Lomniczi A, Loche A, Castellano JM, Ronnekleiv OK, Bosch M, Kaidar G, et al. Epigenetic control of female puberty. Nat Neurosci. 2013;16(3):281-9.
  69. Lomniczi A, Ojeda SR. The Emerging Role of Epigenetics in the Regulation of Female Puberty. Endocr Dev. 2016;29:1-16.
  70. Toro CA, Wright H, Aylwin CF, Ojeda SR, Lomniczi A. Trithorax dependent changes in chromatin landscape at enhancer and promoter regions drive female puberty. Nat Commun. 2018;9(1):57.
  71. Lomniczi A, Wright H, Castellano JM, Matagne V, Toro CA, Ramaswamy S, et al. Epigenetic regulation of puberty via Zinc finger protein-mediated transcriptional repression. Nat Commun. 2015;6:10195.
  72. Barker-Gibb ML, Sahu A, Pohl CR, Plant TM. Elevating circulating leptin in prepubertal male rhesus monkeys (Macaca mulatta) does not elicit precocious gonadotropin-releasing hormone release, assessed indirectly. J Clin Endocrinol Metab. 2002;87(11):4976-83.
  73. Vazquez MJ, Toro CA, Castellano JM, Ruiz-Pino F, Roa J, Beiroa D, et al. SIRT1 mediates obesity- and nutrient-dependent perturbation of pubertal timing by epigenetically controlling Kiss1 expression. Nat Commun. 2018;9(1):4194.
  74. Manfredi-Lozano M, Roa J, Tena-Sempere M. Connecting metabolism and gonadal function: Novel central neuropeptide pathways involved in the metabolic control of puberty and fertility. Front Neuroendocrinol. 2018;48:37-49.
  75. Chachlaki K, Messina A, Delli V, Leysen V, Maurnyi C, Huber C, et al. NOS1 mutations cause hypogonadotropic hypogonadism with sensory and cognitive deficits that can be reversed in infantile mice. Sci Transl Med. 2022;14(665):eabh2369.
  76. Constantin S, Reynolds D, Oh A, Pizano K, Wray S. Nitric oxide resets kisspeptin-excited GnRH neurons via PIP2 replenishment. Proc Natl Acad Sci U S A. 2021;118(1).
  77. Witchel SF, Plant TM. Neurobiology of puberty and its disorders. Handb Clin Neurol. 2021;181:463-96.
  78. Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol. 2022;34(5):e13094.
  79. Terasawa E. The mechanism underlying the pubertal increase in pulsatile GnRH release in primates. J Neuroendocrinol. 2022;34(5):e13119.
  80. Avendano MS, Vazquez MJ, Tena-Sempere M. Disentangling puberty: novel neuroendocrine pathways and mechanisms for the control of mammalian puberty. Hum Reprod Update. 2017;23(6):737-63.
  81. Aylwin CF, Lomniczi A. Sirtuin (SIRT)-1: At the crossroads of puberty and metabolism. Curr Opin Endocr Metab Res. 2020;14:65-72.
  82. Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, et al. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest. 2021;131(18).
  83. Forest MG, Cathiard AM. Pattern of plasma testosterone and delta4-androstenedione in normal newborns: Evidence for testicular activity at birth. J Clin Endocrinol Metab. 1975;41(5):977-80.
  84. Forest MG, Sizonenko PC, Cathiard AM, Bertrand J. Hypophyso-gonadal function in humans during the first year of life. 1. Evidence for testicular activity in early infancy. J Clin Invest. 1974;53(3):819-28.
  85. Grumbach MM. The neuroendocrinology of human puberty revisited. Horm Res. 2002;57 Suppl 2:2-14.
  86. Bizzarri C, Cappa M. Ontogeny of Hypothalamus-Pituitary Gonadal Axis and Minipuberty: An Ongoing Debate? Front Endocrinol (Lausanne). 2020;11:187.
  87. Cortes D, Muller J, Skakkebaek NE. Proliferation of Sertoli cells during development of the human testis assessed by stereological methods. Int J Androl. 1987;10(4):589-96.
  88. Muller J, Skakkebaek NE. Fluctuations in the number of germ cells during the late foetal and early postnatal periods in boys. Acta Endocrinol (Copenh). 1984;105(2):271-4.
  89. Kuijper EA, van Kooten J, Verbeke JI, van Rooijen M, Lambalk CB. Ultrasonographically measured testicular volumes in 0- to 6-year-old boys. Hum Reprod. 2008;23(4):792-6.
  90. Chemes HE, Rey RA, Nistal M, Regadera J, Musse M, Gonzalez-Peramato P, et al. Physiological androgen insensitivity of the fetal, neonatal, and early infantile testis is explained by the ontogeny of the androgen receptor expression in Sertoli cells. J Clin Endocrinol Metab. 2008;93(11):4408-12.
  91. Boukari K, Meduri G, Brailly-Tabard S, Guibourdenche J, Ciampi ML, Massin N, et al. Lack of androgen receptor expression in Sertoli cells accounts for the absence of anti-Mullerian hormone repression during early human testis development. J Clin Endocrinol Metab. 2009;94(5):1818-25.
  92. Codesal J, Regadera J, Nistal M, Regadera-Sejas J, Paniagua R. Involution of human fetal Leydig cells. An immunohistochemical, ultrastructural and quantitative study. J Anat. 1990;172:103-14.
  93. Busch AS, Ljubicic ML, Upners EN, Fischer MB, Raket LL, Frederiksen H, et al. Dynamic Changes of Reproductive Hormones in Male Minipuberty: Temporal Dissociation of Leydig and Sertoli Cell Activity. J Clin Endocrinol Metab. 2022;107(6):1560-8.
  94. Kuiri-Hanninen T, Seuri R, Tyrvainen E, Turpeinen U, Hamalainen E, Stenman UH, et al. Increased activity of the hypothalamic-pituitary-testicular axis in infancy results in increased androgen action in premature boys. J Clin Endocrinol Metab. 2011;96(1):98-105.
  95. Santos MC, Limao S, Ferreira P. Exacerbated mini-puberty of infancy in an ex-extreme preterm girl. BMJ Case Rep. 2020;13(9).
  96. Kuiri-Hanninen T, Kallio S, Seuri R, Tyrvainen E, Liakka A, Tapanainen J, et al. Postnatal developmental changes in the pituitary-ovarian axis in preterm and term infant girls. J Clin Endocrinol Metab. 2011;96(11):3432-9.
  97. Hagen CP, Aksglaede L, Sorensen K, Main KM, Boas M, Cleemann L, et al. Serum levels of anti-Mullerian hormone as a marker of ovarian function in 926 healthy females from birth to adulthood and in 172 Turner syndrome patients. J Clin Endocrinol Metab. 2010;95(11):5003-10.
  98. Ljubicic ML, Busch AS, Upners EN, Fischer MB, Petersen JH, Raket LL, et al. A Biphasic Pattern of Reproductive Hormones in Healthy Female Infants: The COPENHAGEN Minipuberty Study. J Clin Endocrinol Metab. 2022;107(9):2598-605.
  99. Johannsen TH, Main KM, Ljubicic ML, Jensen TK, Andersen HR, Andersen MS, et al. Sex Differences in Reproductive Hormones During Mini-Puberty in Infants With Normal and Disordered Sex Development. J Clin Endocrinol Metab. 2018;103(8):3028-37.
  100. Swee DS, Quinton R. Congenital Hypogonadotrophic Hypogonadism: Minipuberty and the Case for Neonatal Diagnosis. Front Endocrinol (Lausanne). 2019;10:97.
  101. Jespersen K, Ljubicic ML, Johannsen TH, Christiansen P, Skakkebaek NE, Juul A. Distinguishing between hidden testes and anorchia: the role of endocrine evaluation in infancy and childhood. Eur J Endocrinol. 2020;183(1):107-17.
  102. Koo MM, Rohan TE. Accuracy of short-term recall of age at menarche. Ann Hum Biol. 1997;24(1):61-4.
  103. Dorn LD, Sontag-Padilla LM, Pabst S, Tissot A, Susman EJ. Longitudinal reliability of self-reported age at menarche in adolescent girls: variability across time and setting. Dev Psychol. 2013;49(6):1187-93.
  104. Busch AS, Hollis B, Day FR, Sorensen K, Aksglaede L, Perry JRB, et al. Voice break in boys-temporal relations with other pubertal milestones and likely causal effects of BMI. Hum Reprod. 2019;34(8):1514-22.
  105. Lewis ME, Shapland F, Watts R. The influence of chronic conditions and the environment on pubertal development. An example from medieval England. Int J Paleopathol. 2016;12:1-10.
  106. Arthur NA, Gowland RL, Redfern RC. Coming of age in Roman Britain: Osteological evidence for pubertal timing. Am J Phys Anthropol. 2016;159(4):698-713.
  107. Liu W, Yan X, Li C, Shu Q, Chen M, Cai L, et al. A secular trend in age at menarche in Yunnan Province, China: a multiethnic population study of 1,275,000 women. BMC Public Health. 2021;21(1):1890.
  108. Wu T, Pauline M, Germaine BM. Ethnic differences in the Presence of Secondary Sex Characteristics and Menarche Among US Girls:The Third National Health and Nutrition Examination Survey,1988-1994. Pediatrics. 2002;110(4).
  109. Herman-Giddens ME, Slora EJ, Wasserman RC, Bourdony CJ, Bhapkar MV, Koch GG, et al. Secondary sexual characteristics and menses in young girls seen in office practice: a study from the Pediatric Research in Office Settings network. Pediatrics. 1997;99(4):505-12.
  110. Aksglaede L, Sorensen K, Petersen JH, Skakkebaek NE, Juul A. Recent decline in age at breast development: the Copenhagen Puberty Study. Pediatrics. 2009;123(5):e932-9.
  111. Biro FM, Pajak A, Wolff MS, Pinney SM, Windham GC, Galvez MP, et al. Age of Menarche in a Longitudinal US Cohort. J Pediatr Adolesc Gynecol. 2018;31(4):339-45.
  112. Marti-Henneberg C, Vizmanos B. The duration of puberty in girls is related to the timing of its onset. J Pediatr. 1997;131(4):618-21.
  113. Pantsiotou S, Papadimitriou A, Douros K, Priftis K, Nicolaidou P, Fretzayas A. Maturational tempo differences in relation to the timing of the onset of puberty in girls. Acta Paediatr. 2008;97(2):217-20.
  114. Eckert-Lind C, Busch AS, Petersen JH, Biro FM, Butler G, Brauner EV, et al. Worldwide Secular Trends in Age at Pubertal Onset Assessed by Breast Development Among Girls: A Systematic Review and Meta-analysis. JAMA Pediatr. 2020;174(4):e195881.
  115. Sorensen K, Mouritsen A, Aksglaede L, Hagen C, Mogensen S, Juul A. Recent secular Trends in Pubertal Timing: Implications for Evaluation and Diagnosis of Precocious Puberty. Hormone Research in Paediatrics. 2012;77:137-45.
  116. Predieri B, Iughetti L, Bernasconi S, Street ME. Endocrine Disrupting Chemicals' Effects in Children: What We Know and What We Need to Learn? Int J Mol Sci. 2022;23(19).
  117. Krstevska-Konstantinova M, Charlier C, Craen M, Du Caju M, Heinrichs C, de Beaufort C, et al. Sexual precocity after immigration from developing countries to Belgium: evidence of previous exposure to organochlorine pesticides. Hum Reprod. 2001;16(5):1020-6.
  118. Lopez-Rodriguez D, Franssen D, Heger S, Parent AS. Endocrine-disrupting chemicals and their effects on puberty. Best Pract Res Clin Endocrinol Metab. 2021;35(5):101579.
  119. Fudvoye J, Lopez-Rodriguez D, Franssen D, Parent AS. Endocrine disrupters and possible contribution to pubertal changes. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101300.
  120. Kaplowitz PB, Slora EJ, Wasserman RC, Pedlow SE, Herman-Giddens ME. Earlier onset of puberty in girls: relation to increased body mass index and race. Pediatrics. 2001;108(2):347-53.
  121. Kaplowitz PB. Link between body fat and the timing of puberty. Pediatrics. 2008;121 Suppl 3:S208-17.
  122. Brix N, Ernst A, Lauridsen LLB, Parner ET, Arah OA, Olsen J, et al. Childhood overweight and obesity and timing of puberty in boys and girls: cohort and sibling-matched analyses. Int J Epidemiol. 2020;49(3):834-44.
  123. Ahmed ML, Ong KK, Dunger DB. Childhood obesity and the timing of puberty. Trends Endocrinol Metab. 2009;20(5):237-42.
  124. Day FR, Perry JR, Ong KK. Genetic Regulation of Puberty Timing in Humans. Neuroendocrinology. 2015;102(4):247-55.
  125. Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr. 2022;95(2):120-36.
  126. Reid RL, Ling N, Yen SS. Gonadotropin-releasing activity of alpha-melanocyte-stimulating hormone in normal subjects and in subjects with hypothalamic-pituitary dysfunction. J Clin Endocrinol Metab. 1984;58(5):773-7.
  127. Israel DD, Sheffer-Babila S, de Luca C, Jo YH, Liu SM, Xia Q, et al. Effects of leptin and melanocortin signaling interactions on pubertal development and reproduction. Endocrinology. 2012;153(5):2408-19.
  128. Morris DH, Jones ME, Schoemaker MJ, Ashworth A, Swerdlow AJ. Familial concordance for age at menarche: analyses from the Breakthrough Generations Study. Paediatr Perinat Epidemiol. 2011;25(3):306-11.
  129. Palmert MR, Hirschhorn JN. Genetic approaches to stature, pubertal timing, and other complex traits. Mol Genet Metab. 2003;80(1-2):1-10.
  130. Busch AS, Hagen CP, Juul A. Heritability of pubertal timing: detailed evaluation of specific milestones in healthy boys and girls. Eur J Endocrinol. 2020;183(1):13-20.
  131. Wohlfahrt-Veje C, Mouritsen A, Hagen CP, Tinggaard J, Mieritz MG, Boas M, et al. Pubertal Onset in Boys and Girls Is Influenced by Pubertal Timing of Both Parents. J Clin Endocrinol Metab. 2016;101(7):2667-74.
  132. Day FR, Thompson DJ, Helgason H, Chasman DI, Finucane H, Sulem P, et al. Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk. Nat Genet. 2017;49(6):834-41.
  133. Sarnowski C, Cousminer DL, Franceschini N, Raffield LM, Jia G, Fernandez-Rhodes L, et al. Large trans-ethnic meta-analysis identifies AKR1C4 as a novel gene associated with age at menarche. Hum Reprod. 2021;36(7):1999-2010.
  134. Klein KO. Precocious puberty: who has it? Who should be treated? J Clin Endocrinol Metab. 1999;84(2):411-4.
  135. Parent AS, Teilmann G, Juul A, Skakkebaek NE, Toppari J, Bourguignon JP. The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration. Endocr Rev. 2003;24(5):668-93.
  136. Kaplowitz PB. Delayed puberty. Pediatr Rev. 2010;31(5):189-95.
  137. Bangalore Krishna K, Fuqua JS, Rogol AD, Klein KO, Popovic J, Houk CP, et al. Use of Gonadotropin-Releasing Hormone Analogs in Children: Update by an International Consortium. Horm Res Paediatr. 2019;91(6):357-72.
  138. Goldberg M, D'Aloisio AA, O'Brien KM, Zhao S, Sandler DP. Pubertal timing and breast cancer risk in the Sister Study cohort. Breast Cancer Res. 2020;22(1):112.
  139. Mueller NT, Duncan BB, Barreto SM, Chor D, Bessel M, Aquino EML, et al. Earlier age at menarche is associated with higher diabetes risk and cardiometabolic disease risk factors in Brazilian adults: Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). Cardiovasc Diabetol. 2014;13.
  140. Partsch CJ, Heger S, Sippell WG. Management and outcome of central precocious puberty. Clin Endocrinol (Oxf). 2002;56(2):129-48.
  141. Soriano-Guillen L, Corripio R, Labarta JI, Canete R, Castro-Feijoo L, Espino R, et al. Central precocious puberty in children living in Spain: incidence, prevalence, and influence of adoption and immigration. J Clin Endocrinol Metab. 2010;95(9):4305-13.
  142. Kim SH, Huh K, Won S, Lee KW, Park MJ. A Significant Increase in the Incidence of Central Precocious Puberty among Korean Girls from 2004 to 2010. Plos One. 2015;10(11).
  143. Brandberg G, Raininko R, Eeg-Olofsson O. Hypothalamic hamartoma with gelastic seizures in Swedish children and adolescents. Eur J Paediatr Neurol. 2004;8(1):35-44.
  144. Cukier P, Castro LH, Banaskiwitz N, Teles LR, Ferreira LR, Adda CC, et al. The benign spectrum of hypothalamic hamartomas: infrequent epilepsy and normal cognition in patients presenting with central precocious puberty. Seizure. 2013;22(1):28-32.
  145. Harrison VS, Oatman O, Kerrigan JF. Hypothalamic hamartoma with epilepsy: Review of endocrine comorbidity. Epilepsia. 2017;58 Suppl 2(Suppl 2):50-9.
  146. Yang Y, Shen F, Jing XP, Zhang N, Xu SY, Li DD, et al. Case Report: Whole-Exome Sequencing of Hypothalamic Hamartoma From an Infant With Pallister-Hall Syndrome Revealed Novel de novo Mutation in the GLI3. Front Surg. 2021;8:734757.
  147. Fujita A, Higashijima T, Shirozu H, Masuda H, Sonoda M, Tohyama J, et al. Pathogenic variants of DYNC2H1, KIAA0556, and PTPN11 associated with hypothalamic hamartoma. Neurology. 2019;93(3):e237-e51.
  148. Hildebrand MS, Griffin NG, Damiano JA, Cops EJ, Burgess R, Ozturk E, et al. Mutations of the Sonic Hedgehog Pathway Underlie Hypothalamic Hamartoma with Gelastic Epilepsy. Am J Hum Genet. 2016;99(2):423-9.
  149. Chan YM, Fenoglio-Simeone KA, Paraschos S, Muhammad L, Troester MM, Ng YT, et al. Central precocious puberty due to hypothalamic hamartomas correlates with anatomic features but not with expression of GnRH, TGFalpha, or KISS1. Horm Res Paediatr. 2010;73(5):312-9.
  150. Harrison VS, Oatman O, Kerrigan JF. Hypothalamic hamartoma with epilepsy: Review of endocrine comorbidity. Epilepsia. 2017;58:50-9.
  151. Bourdillon P, Ferrand-Sorbet S, Apra C, Chipaux M, Raffo E, Rosenberg S, et al. Surgical treatment of hypothalamic hamartomas. Neurosurg Rev. 2021;44(2):753-62.
  152. Chalumeau M, Chemaitilly W, Trivin C, Adan L, Breart G, Brauner R. Central precocious puberty in girls: an evidence-based diagnosis tree to predict central nervous system abnormalities. Pediatrics. 2002;109(1):61-7.
  153. Stephen MD, Zage PE, Waguespack SG. Gonadotropin-dependent precocious puberty: neoplastic causes and endocrine considerations. Int J Pediatr Endocrinol. 2011;2011(1):184502.
  154. Pinheiro SL, Maciel J, Cavaco D, Figueiredo AA, Damasio IL, Donato S, et al. Precocious and accelerated puberty in children with neurofibromatosis type 1: results from a close follow-up of a cohort of 45 patients. Hormones. 2023;22(1):79-85.
  155. Almutlaq N, O'Neil J, Fuqua JS. Central precocious puberty in spina bifida children: Guidelines for the care of people with spina bifida. J Pediatr Rehabil Med. 2020;13(4):557-63.
  156. Cerbone M, Guemes M, Wade A, Improda N, Dattani M. Endocrine morbidity in midline brain defects: Differences between septo-optic dysplasia and related disorders. EClinicalMedicine. 2020;19:100224.
  157. Oatman OJ, McClellan DR, Olson ML, Garcia-Filion P. Endocrine and pubertal disturbances in optic nerve hypoplasia, from infancy to adolescence. Int J Pediatr Endocrinol. 2015;2015(1):8.
  158. Siegel DA, King JB, Lupo PJ, Durbin EB, Tai E, Mills K, et al. Counts, incidence rates, and trends of pediatric cancer in the United States, 2003-2019. J Natl Cancer Inst. 2023.
  159. Chemaitilly W, Merchant TE, Li Z, Barnes N, Armstrong GT, Ness KK, et al. Central precocious puberty following the diagnosis and treatment of paediatric cancer and central nervous system tumours: presentation and long-term outcomes. Clin Endocrinol (Oxf). 2016;84(3):361-71.
  160. De Sanctis V, Soliman AT, Elsedfy H, Soliman NA, Elalaily R, El Kholy M. Precocious Puberty Following Traumatic Brain Injury in Early Childhood: A Review of the Literature. Pediatr Endocrinol Rev. 2015;13(1):458-64.
  161. Dassa Y, Crosnier H, Chevignard M, Viaud M, Personnier C, Flechtner I, et al. Pituitary deficiency and precocious puberty after childhood severe traumatic brain injury: a long-term follow-up prospective study. Eur J Endocrinol. 2019;180(5):281-90.
  162. Partsch CJ, Sippell WG. Pathogenesis and epidemiology of precocious puberty. Effects of exogenous oestrogens. Hum Reprod Update. 2001;7(3):292-302.
  163. Stecchini MF, Braid Z, More CB, Aragon DC, Castro M, Moreira AC, et al. Gonadotropin-dependent pubertal disorders are common in patients with virilizing adrenocortical tumors in childhood. Endocr Connect. 2019;8(5):579-89.
  164. Chen H, Mo CY, Zhong LY. Central precocious puberty secondary to peripheral precocious puberty due to a pineal germ cell tumor: a case and review of literature. BMC Endocr Disord. 2023;23(1):237.
  165. Maione L, Bouvattier C, Kaiser UB. Central precocious puberty: Recent advances in understanding the aetiology and in the clinical approach. Clin Endocrinol (Oxf). 2021;95(4):542-55.
  166. Moise-Silverman J, Silverman LA. A review of the genetics and epigenetics of central precocious puberty. Front Endocrinol (Lausanne). 2022;13:1029137.
  167. Valadares LP, Meireles CG, De Toledo IP, Santarem de Oliveira R, Goncalves de Castro LC, Abreu AP, et al. MKRN3 Mutations in Central Precocious Puberty: A Systematic Review and Meta-Analysis. J Endocr Soc. 2019;3(5):979-95.
  168. Magnotto JC, Mancini A, Bird K, Montenegro L, Tutunculer F, Pereira SA, et al. Novel MKRN3 Missense Mutations Associated With Central Precocious Puberty Reveal Distinct Effects on Ubiquitination. J Clin Endocrinol Metab. 2023;108(7):1646-56.
  169. Li M, Chen Y, Liao B, Tang J, Zhong J, Lan D. The role of kisspeptin and MKRN3 in the diagnosis of central precocious puberty in girls. Endocr Connect. 2021;10(9):1147-54.
  170. Ioannides Y, Lokulo-Sodipe K, Mackay DJ, Davies JH, Temple IK. Temple syndrome: improving the recognition of an underdiagnosed chromosome 14 imprinting disorder: an analysis of 51 published cases. J Med Genet. 2014;51(8):495-501.
  171. Gomes LG, Cunha-Silva M, Crespo RP, Ramos CO, Montenegro LR, Canton A, et al. DLK1 Is a Novel Link Between Reproduction and Metabolism. J Clin Endocrinol Metab. 2019;104(6):2112-20.
  172. Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, et al. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276-80.
  173. Teles MG, Bianco SD, Brito VN, Trarbach EB, Kuohung W, Xu S, et al. A GPR54-activating mutation in a patient with central precocious puberty. N Engl J Med. 2008;358(7):709-15.
  174. Bianco SD, Vandepas L, Correa-Medina M, Gereben B, Mukherjee A, Kuohung W, et al. KISS1R intracellular trafficking and degradation: effect of the Arg386Pro disease-associated mutation. Endocrinology. 2011;152(4):1616-26.
  175. Tinano FR, Canton APM, Montenegro LR, de Castro Leal A, Faria AG, Seraphim CE, et al. Clinical and Genetic Characterization of Familial Central Precocious Puberty. J Clin Endocrinol Metab. 2023;108(7):1758-67.
  176. Nicoara DM, Scutca AC, Mang N, Juganaru I, Munteanu AI, Vitan L, et al. Central precocious puberty in Prader-Willi syndrome: a narrative review. Front Endocrinol (Lausanne). 2023;14:1150323.
  177. Spielmann S, Partsch CJ, Gosch A, Pankau R. Treatment of central precocious puberty and early puberty with GnRH analog in girls with Williams-Beuren syndrome. J Pediatr Endocrinol Metab. 2015;28(11-12):1363-7.
  178. Patti G, Malerba F, Calevo MG, Schiavone M, Scaglione M, Casalini E, et al. Pubertal timing in children with Silver Russell syndrome compared to those born small for gestational age. Front Endocrinol (Lausanne). 2022;13:975511.
  179. Palmert MR, Malin HV, Boepple PA. Unsustained or slowly progressive puberty in young girls: initial presentation and long-term follow-up of 20 untreated patients. J Clin Endocrinol Metab. 1999;84(2):415-23.
  180. Cote DJ, Smith TR, Sandler CN, Gupta T, Bale TA, Bi WL, et al. Functional Gonadotroph Adenomas: Case Series and Report of Literature. Neurosurgery. 2016;79(6):823-31.
  181. Ntali G, Capatina C. Updating the Landscape for Functioning Gonadotroph Tumors. Medicina (Kaunas). 2022;58(8).
  182. Uhing A, Ahmed A, Salamat S, Chen M. A Rare Case of Precocious Puberty Secondary to an LH-secreting Pituitary Adenoma. JCEM Case Rep. 2023;1(3):luad055.
  183. Sisk-Hackworth L, Kelley ST, Thackray VG. Sex, puberty, and the gut microbiome. Reproduction. 2023;165(2):R61-R74.
  184. Calcaterra V, Rossi V, Massini G, Regalbuto C, Hruby C, Panelli S, et al. Precocious puberty and microbiota: The role of the sex hormone-gut microbiome axis. Front Endocrinol (Lausanne). 2022;13:1000919.
  185. Chaussain JL, Roger M, Couprie C, Lahlou N, Canlorbe P. Treatment of precocious puberty with a long-acting preparation of D-Trp6-LHRH. Horm Res. 1987;28(2-4):155-63.
  186. Lahlou N, Roger M, Chaussain JL, Feinstein MC, Sultan C, Toublanc JE, et al. Gonadotropin and alpha-subunit secretion during long term pituitary suppression by D-Trp6-luteinizing hormone-releasing hormone microcapsules as treatment of precocious puberty. J Clin Endocrinol Metab. 1987;65(5):946-53.
  187. Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2366-77.
  188. Chen M, Eugster EA. Central Precocious Puberty: Update on Diagnosis and Treatment. Pediatr Drugs. 2015;17(4):273-81.
  189. Johnson SR, Nolan RC, Grant MT, Price GJ, Siafarikas A, Bint L, et al. Sterile abscess formation associated with depot leuprorelin acetate therapy for central precocious puberty. J Paediatr Child Health. 2012;48(3):E136-9.
  190. Miller BS, Shukla AR. Sterile abscess formation in response to two separate branded long-acting gonadotropin-releasing hormone agonists. Clin Ther. 2010;32(10):1749-51.
  191. Pienkowski C, Tauber M. Gonadotropin-Releasing Hormone Agonist Treatment in Sexual Precocity. Endocr Dev. 2016;29:214-29.
  192. De Sanctis V, Soliman AT, Di Maio S, Soliman N, Elsedfy H. Long-term effects and significant Adverse Drug Reactions (ADRs) associated with the use of Gonadotropin-Releasing Hormone analogs (GnRHa) for central precocious puberty: a brief review of literature. Acta Biomed. 2019;90(3):345-59.
  193. van der Sluis IM, Boot AM, Krenning EP, Drop SL, de Muinck Keizer-Schrama SM. Longitudinal follow-up of bone density and body composition in children with precocious or early puberty before, during and after cessation of GnRH agonist therapy. J Clin Endocrinol Metab. 2002;87(2):506-12.
  194. Paterson WF, McNeill E, Young D, Donaldson MD. Auxological outcome and time to menarche following long-acting goserelin therapy in girls with central precocious or early puberty. Clin Endocrinol (Oxf). 2004;61(5):626-34.
  195. Traggiai C, Perucchin PP, Zerbini K, Gastaldi R, De Biasio P, Lorini R. Outcome after depot gonadotrophin-releasing hormone agonist treatment for central precocious puberty: effects on body mass index and final height. Eur J Endocrinol. 2005;153(3):463-4.
  196. Wolters B, Lass N, Reinehr T. Treatment with gonadotropin-releasing hormone analogues: different impact on body weight in normal-weight and overweight children. Horm Res Paediatr. 2012;78(5-6):304-11.
  197. Palmert MR, Mansfield MJ, Crowley WF, Crigler JF, Crawford JD, Boepple PA. Is obesity an outcome of gonadotropin-releasing hormone agonist administration? Analysis of growth and body composition in 110 patients with central precocious puberty. J Clin Endocrinol Metab. 1999;84(12):4480-8.
  198. Boot AM, De Muinck Keizer-Schrama S, Pols HA, Krenning EP, Drop SL. Bone mineral density and body composition before and during treatment with gonadotropin-releasing hormone agonist in children with central precocious and early puberty. J Clin Endocrinol Metab. 1998;83(2):370-3.
  199. Lazar L, Lebenthal Y, Yackobovitch-Gavan M, Shalitin S, de Vries L, Phillip M, et al. Treated and untreated women with idiopathic precocious puberty: BMI evolution, metabolic outcome, and general health between third and fifth decades. J Clin Endocrinol Metab. 2015;100(4):1445-51.
  200. Guaraldi F, Beccuti G, Gori D, Ghizzoni L. MANAGEMENT OF ENDOCRINE DISEASE: Long-term outcomes of the treatment of central precocious puberty. Eur J Endocrinol. 2016;174(3):R79-87.
  201. Lee JW, Kim HJ, Choe YM, Kang HS, Kim SK, Jun YH, et al. Significant adverse reactions to long-acting gonadotropin-releasing hormone agonists for the treatment of central precocious puberty and early onset puberty. Ann Pediatr Endocrinol Metab. 2014;19(3):135-40.
  202. Omar AA, Nyaga G, Mungai LNW. Pseudotumor cerebri in patient on leuprolide acetate for central precocious puberty. Int J Pediatr Endocrinol. 2020;2020(1):22.
  203. Lazar L, Meyerovitch J, de Vries L, Phillip M, Lebenthal Y. Treated and untreated women with idiopathic precocious puberty: long-term follow-up and reproductive outcome between the third and fifth decades. Clin Endocrinol (Oxf). 2014;80(4):570-6.
  204. Bertelloni S, Baroncelli GI, Ferdeghini M, Menchini-Fabris F, Saggese G. Final height, gonadal function and bone mineral density of adolescent males with central precocious puberty after therapy with gonadotropin-releasing hormone analogues. Eur J Pediatr. 2000;159(5):369-74.
  205. Thornton P, Silverman LA, Geffner ME, Neely EK, Gould E, Danoff TM. Review of outcomes after cessation of gonadotropin-releasing hormone agonist treatment of girls with precocious puberty. Pediatr Endocrinol Rev. 2014;11(3):306-17.
  206. Arcari AJ, Freire AV, Ballerini MG, Escobar ME, Diaz Marsiglia YM, Bergada I, et al. Prevalence of Polycystic Ovarian Syndrome in Girls with a History of Idiopathic Central Precocious Puberty. Horm Res Paediatr. 2023:1-6.
  207. Eugster EA. Treatment of Central Precocious Puberty. J Endocr Soc. 2019;3(5):965-72.
  208. Schoelwer MJ, Donahue KL, Didrick P, Eugster EA. One-Year Follow-Up of Girls with Precocious Puberty and Their Mothers: Do Psychological Assessments Change over Time or with Treatment? Horm Res Paediatr. 2017;88(5):347-53.
  209. Kaplowitz PB, Backeljauw PF, Allen DB. Toward More Targeted and Cost-Effective Gonadotropin-Releasing Hormone Analog Treatment in Girls with Central Precocious Puberty. Horm Res Paediatr. 2018;90(1):1-7.
  210. Popovic J, Geffner ME, Rogol AD, Silverman LA, Kaplowitz PB, Mauras N, et al. Gonadotropin-releasing hormone analog therapies for children with central precocious puberty in the United States. Frontiers in Pediatrics. 2022;10.
  211. Eugster EA. Treatment of Central Precocious Puberty. Journal of the Endocrine Society. 2019;3(5):965-72.
  212. Pasquino AM, Municchi G, Pucarelli I, Segni M, Mancini MA, Troiani S. Combined treatment with gonadotropin-releasing hormone analog and growth hormone in central precocious puberty. J Clin Endocrinol Metab. 1996;81(3):948-51.
  213. Shi Y, Ma Z, Yang X, Ying Y, Luo X, Hou L. Gonadotropin-releasing hormone analogue and recombinant human growth hormone treatment for idiopathic central precocious puberty in girls. Front Endocrinol (Lausanne). 2022;13:1085385.
  214. Cho AY, Shim YS, Lee HS, Hwang JS. Effect of gonadotropin-releasing hormone agonist monotherapy and combination therapy with growth hormone on final adult height in girls with central precocious puberty. Sci Rep. 2023;13(1):1264.
  215. Pucarelli I, Segni M, Ortore M, Arcadi E, Pasquino AM. Effects of combined gonadotropin-releasing hormone agonist and growth hormone therapy on adult height in precocious puberty: a further contribution. J Pediatr Endocrinol Metab. 2003;16(7):1005-10.
  216. Li YH, Zhu SY, Ma HM, Su Z, Chen HS, Chen QL, et al. [Effect of gonadotropin-releasing hormone analog combined with stanazolol on final height in girls with idiopathic central precocious puberty and apparent decrease of linear growth]. Zhonghua Er Ke Za Zhi. 2013;51(11):807-12.
  217. Zhu S, Long L, Hu Y, Tuo Y, Li Y, Yu Z. GnRHa/Stanozolol Combined Therapy Maintains Normal Bone Growth in Central Precocious Puberty. Front Endocrinol (Lausanne). 2021;12:678797.
  218. Vottero A, Pedori S, Verna M, Pagano B, Cappa M, Loche S, et al. Final height in girls with central idiopathic precocious puberty treated with gonadotropin-releasing hormone analog and oxandrolone. J Clin Endocrinol Metab. 2006;91(4):1284-7.
  219. Liu S, Liu Q, Cheng X, Luo Y, Wen Y. Effects and safety of combination therapy with gonadotropin-releasing hormone analogue and growth hormone in girls with idiopathic central precocious puberty: a meta-analysis. J Endocrinol Invest. 2016;39(10):1167-78.
  220. Wit JM. Should Skeletal Maturation Be Manipulated for Extra Height Gain? Front Endocrinol (Lausanne). 2021;12:812196.
  221. Dotremont H, France A, Heinrichs C, Tenoutasse S, Brachet C, Cools M, et al. Efficacy and safety of a 4-year combination therapy of growth hormone and gonadotropin-releasing hormone analogue in pubertal girls with short predicted adult height. Front Endocrinol (Lausanne). 2023;14:1113750.
  222. Trujillo MV, Lee PA, Reifschneider K, Backeljauw PF, Dragnic S, Van Komen S, et al. Using change in predicted adult height during GnRH agonist treatment for individualized treatment decisions in girls with central precocious puberty. J Pediatr Endocrinol Metab. 2023;36(3):299-308.
  223. Vargas Trujillo M, Dragnic S, Aldridge P, Klein KO. Importance of individualizing treatment decisions in girls with central precocious puberty when initiating treatment after age 7 years or continuing beyond a chronological age of 10 years or a bone age of 12 years. J Pediatr Endocrinol Metab. 2021;34(6):733-9.
  224. Mendle J, Ryan RM, McKone KMP. Age at Menarche, Depression, and Antisocial Behavior in Adulthood. Pediatrics. 2018;141(1).
  225. Mendle J, Ryan RM, McKone KMP. Early Menarche and Internalizing and Externalizing in Adulthood: Explaining the Persistence of Effects. J Adolescent Health. 2019;65(5):599-606.
  226. Schoelwer MJ, Donahue KL, Didrick P, Eugster EA. One-Year Follow-Up of Girls with Precocious Puberty and Their Mothers: Do Psychological Assessments Change over Time or with Treatment? Hormone Research in Paediatrics. 2017;88(5):347-53.
  227. Wojniusz S, Callens N, Sütterlin S, Andersson S, De Schepper J, Gies I, et al. Cognitive, Emotional, and Psychosocial Functioning of Girls Treated with Pharmacological Puberty Blockage for Idiopathic Central Precocious Puberty. Front Psychol. 2016;7:1053.
  228. Williams VSL, Soliman AM, Barrett AM, Klein KO. Review and evaluation of patient-centered psychosocial assessments for children with central precocious puberty or early puberty. J Pediatr Endocrinol Metab. 2018;31(5):485-95.
  229. Lewis KA, Eugster EA. Random luteinizing hormone often remains pubertal in children treated with the histrelin implant for central precocious puberty. J Pediatr. 2013;162(3):562-5.
  230. Neely EK, Silverman LA, Geffner ME, Danoff TM, Gould E, Thornton PS. Random unstimulated pediatric luteinizing hormone levels are not reliable in the assessment of pubertal suppression during histrelin implant therapy. Int J Pediatr Endocrinol. 2013;2013(1):20.
  231. Klein KO, Lee PA. Gonadotropin-Releasing Hormone (GnRHa) Therapy for Central Precocious Puberty (CPP): Review of Nuances in Assessment of Height, Hormonal Suppression, Psychosocial Issues, and Weight Gain, with Patient Examples. Pediatr Endocrinol Rev. 2018;15(4):298-312.
  232. Houk CP, Kunselman AR, Lee PA. The diagnostic value of a brief GnRH analogue stimulation test in girls with central precocious puberty: a single 30-minute post-stimulation LH sample is adequate. J Pediatr Endocrinol Metab. 2008;21(12):1113-8.
  233. Yazdani P, Lin Y, Raman V, Haymond M. A single sample GnRHa stimulation test in the diagnosis of precocious puberty. Int J Pediatr Endocrinol. 2012;2012(1):23.
  234. Ohyama K, Tanaka T, Tachibana K, Niimi H, Fujieda K, Matsuo N, et al. Timing for discontinuation of treatment with a long-acting gonadotropin-releasing hormone analog in girls with central precocious puberty. TAP-144SR CPP Study Group. Endocr J. 1998;45(3):351-6.
  235. Carel JC, Roger M, Ispas S, Tondu F, Lahlou N, Blumberg J, et al. Final height after long-term treatment with triptorelin slow release for central precocious puberty: importance of statural growth after interruption of treatment. French study group of Decapeptyl in Precocious Puberty. J Clin Endocrinol Metab. 1999;84(6):1973-8.
  236. Carel JC, Eugster EA, Rogol A, Ghizzoni L, Palmert MR, Antoniazzi F, et al. Consensus statement on the use of gonadotropin-releasing hormone analogs in children. Pediatrics. 2009;123(4):e752-62.
  237. Boyce AM, Collins MT. Fibrous Dysplasia/McCune-Albright Syndrome: A Rare, Mosaic Disease of Galpha s Activation. Endocr Rev. 2020;41(2):345-70.
  238. Tufano M, Ciofi D, Amendolea A, Stagi S. Auxological and Endocrinological Features in Children With McCune Albright Syndrome: A Review. Front Endocrinol (Lausanne). 2020;11:522.
  239. Spencer T, Pan KS, Collins MT, Boyce AM. The Clinical Spectrum of McCune-Albright Syndrome and Its Management. Horm Res Paediatr. 2019;92(6):347-56.
  240. Kim IS, Kim ER, Nam HJ, Chin MO, Moon YH, Oh MR, et al. Activating mutation of GS alpha in McCune-Albright syndrome causes skin pigmentation by tyrosinase gene activation on affected melanocytes. Horm Res. 1999;52(5):235-40.
  241. Szymczuk V, Taylor J, Michel Z, Sinaii N, Boyce AM. Skeletal Disease Acquisition in Fibrous Dysplasia: Natural History and Indicators of Lesion Progression in Children. J Bone Miner Res. 2022;37(8):1473-8.
  242. Collins MT, Chebli C, Jones J, Kushner H, Consugar M, Rinaldo P, et al. Renal phosphate wasting in fibrous dysplasia of bone is part of a generalized renal tubular dysfunction similar to that seen in tumor-induced osteomalacia. J Bone Miner Res. 2001;16(5):806-13.
  243. Bhattacharyya N, Wiench M, Dumitrescu C, Connolly BM, Bugge TH, Patel HV, et al. Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner Res. 2012;27(5):1132-41.
  244. Boyce AM, Casey RK, Ovejero Crespo D, Murdock CM, Estrada A, Guthrie LC, et al. Gynecologic and reproductive outcomes in fibrous dysplasia/McCune-Albright syndrome. Orphanet J Rare Dis. 2019;14(1):90.
  245. Boyce AM, Chong WH, Shawker TH, Pinto PA, Linehan WM, Bhattacharryya N, et al. Characterization and management of testicular pathology in McCune-Albright syndrome. J Clin Endocrinol Metab. 2012;97(9):E1782-90.
  246. Tatsi C, Stratakis CA. Neonatal Cushing Syndrome: A Rare but Potentially Devastating Disease. Clin Perinatol. 2018;45(1):103-18.
  247. Gaujoux S, Salenave S, Ronot M, Rangheard AS, Cros J, Belghiti J, et al. Hepatobiliary and Pancreatic neoplasms in patients with McCune-Albright syndrome. J Clin Endocrinol Metab. 2014;99(1):E97-101.
  248. Zacharin M, Bajpai A, Chow CW, Catto-Smith A, Stratakis C, Wong MW, et al. Gastrointestinal polyps in McCune Albright syndrome. J Med Genet. 2011;48(7):458-61.
  249. Majoor BC, Boyce AM, Bovee JV, Smit VT, Collins MT, Cleton-Jansen AM, et al. Increased Risk of Breast Cancer at a Young Age in Women with Fibrous Dysplasia. J Bone Miner Res. 2018;33(1):84-90.
  250. Lumbroso S, Paris F, Sultan C, European Collaborative S. Activating Gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome--a European Collaborative Study. J Clin Endocrinol Metab. 2004;89(5):2107-13.
  251. Roszko KL, Guthrie L, Li X, Collins MT, de Castro LF, Boyce AM. Identification of GNAS Variants in Circulating Cell-Free DNA from Patients with Fibrous Dysplasia/McCune Albright Syndrome. J Bone Miner Res. 2023;38(3):443-50.
  252. Couch RM, Muller J, Perry YS, Winter JS. Kinetic analysis of inhibition of human adrenal steroidogenesis by ketoconazole. J Clin Endocrinol Metab. 1987;65(3):551-4.
  253. Estrada A, Boyce AM, Brillante BA, Guthrie LC, Gafni RI, Collins MT. Long-term outcomes of letrozole treatment for precocious puberty in girls with McCune-Albright syndrome. Eur J Endocrinol. 2016;175(5):477-83.
  254. Eugster EA, Rubin SD, Reiter EO, Plourde P, Jou HC, Pescovitz OH, et al. Tamoxifen treatment for precocious puberty in McCune-Albright syndrome: a multicenter trial. J Pediatr. 2003;143(1):60-6.
  255. Hu R, Hilakivi-Clarke L, Clarke R. Molecular mechanisms of tamoxifen-associated endometrial cancer (Review). Oncol Lett. 2015;9(4):1495-501.
  256. Sims EK, Garnett S, Guzman F, Paris F, Sultan C, Eugster EA, et al. Fulvestrant treatment of precocious puberty in girls with McCune-Albright syndrome. Int J Pediatr Endocrinol. 2012;2012(1):26.
  257. Nabhan ZM, West KW, Eugster EA. Oophorectomy in McCune-Albright syndrome: a case of mistaken identity. J Pediatr Surg. 2007;42(9):1578-83.
  258. Corica D, Aversa T, Pepe G, De Luca F, Wasniewska M. Peculiarities of Precocious Puberty in Boys and Girls With McCune-Albright Syndrome. Front Endocrinol (Lausanne). 2018;9:337.
  259. Tessaris D, Matarazzo P, Mussa A, Tuli G, Verna F, Fiore L, et al. Combined treatment with bicalutamide and anastrozole in a young boy with peripheral precocious puberty due to McCune-Albright Syndrome. Endocr J. 2012;59(2):111-7.
  260. Partsch CJ, Kreller-Laugwitz G, Sippell WG. [Transitory precocious puberty caused by autonomous ovarian cysts. Clinical, endocrinologic and sonographic course]. Monatsschr Kinderheilkd. 1989;137(4):235-8.
  261. de Sousa G, Wunsch R, Andler W. Precocious pseudopuberty due to autonomous ovarian cysts: a report of ten cases and long-term follow-up. Hormones. 2008;7(2):170-4.
  262. Nayak S, Witchel SF, Sanfilippo JS. Vaginal foreign body: a delayed diagnosis. J Pediatr Adolesc Gynecol. 2014;27(6):e127-9.
  263. Ganer Herman H, Shalev A, Ginat S, Kerner R, Keidar R, Bar J, et al. Clinical characteristics of adnexal torsion in premenarchal patients. Arch Gynecol Obstet. 2016;293(3):603-8.
  264. Adeyemi-Fowode O, Lin EG, Syed F, Sangi-Haghpeykar H, Zhu H, Dietrich JE. Adnexal Torsion in Children and Adolescents: A Retrospective Review of 245 Cases at a Single Institution. J Pediatr Adolesc Gynecol. 2019;32(1):64-9.
  265. Spinelli C, Piscioneri J, Strambi S. Adnexal torsion in adolescents: update and review of the literature. Curr Opin Obstet Gynecol. 2015;27(5):320-5.
  266. Spinelli C, Buti I, Pucci V, Liserre J, Alberti E, Nencini L, et al. Adnexal torsion in children and adolescents: new trends to conservative surgical approach -- our experience and review of literature. Gynecol Endocrinol. 2013;29(1):54-8.
  267. Heo S, Shim YS, Lee HS, Hwang JS. Clinical course of peripheral precocious puberty in girls due to autonomous ovarian cysts. Clin Endocrinol (Oxf). 2023.
  268. Poonai N, Poonai C, Lim R, Lynch T. Pediatric ovarian torsion: case series and review of the literature. Can J Surg. 2013;56(2):103-8.
  269. Gaikwad PM, Goswami S, Sengupta N, Baidya A, Das N. Transformation of Peripheral Sexual Precocity to Central Sexual Precocity Following Treatment of Granulosa Cell Tumor of the Ovary. Cureus. 2022;14(2):e22676.
  270. Bessiere L, Todeschini AL, Auguste A, Sarnacki S, Flatters D, Legois B, et al. A Hot-spot of In-frame Duplications Activates the Oncoprotein AKT1 in Juvenile Granulosa Cell Tumors. EBioMedicine. 2015;2(5):421-31.
  271. Fuller PJ, Leung D, Chu S. Genetics and genomics of ovarian sex cord-stromal tumors. Clin Genet. 2017;91(2):285-91.
  272. Yamamoto H, Sakamoto H, Kumagai H, Abe T, Ishiguro S, Uchida K, et al. Clinical Guidelines for Diagnosis and Management of Peutz-Jeghers Syndrome in Children and Adults. Digestion. 2023:1-13.
  273. De Paolis E, Paragliola RM, Concolino P. Spectrum of DICER1 Germline Pathogenic Variants in Ovarian Sertoli-Leydig Cell Tumor. J Clin Med. 2021;10(9).
  274. Patel SS, Carrick KS, Carr BR. Virilization persists in a woman with an androgen-secreting granulosa cell tumor. Fertil Steril. 2009;91(3):933 e13-5.
  275. Kalfa N, Meduri G, Philibert P, Patte C, Boizet-Bonhoure B, Thibaut E, et al. Unusual virilization in girls with juvenile granulosa cell tumors of the ovary is related to intratumoral aromatase deficiency. Horm Res Paediatr. 2010;74(2):83-91.
  276. Schultz KAP, Williams GM, Kamihara J, Stewart DR, Harris AK, Bauer AJ, et al. DICER1 and Associated Conditions: Identification of At-risk Individuals and Recommended Surveillance Strategies. Clin Cancer Res. 2018;24(10):2251-61.
  277. Trahmono, Wahyudi I, Rodjani A, Situmorang GR, Marzuki NS. Precocious Pseudo-Puberty with Testicular Enlargement: Two Cases of Leydig Cell Tumor with Different Histopathological Results. Res Rep Urol. 2020;12:577-82.
  278. Luckie TM, Danzig M, Zhou S, Wu H, Cost NG, Karaviti L, et al. A Multicenter Retrospective Review of Pediatric Leydig Cell Tumor of the Testis. J Pediatr Hematol Oncol. 2019;41(1):74-6.
  279. De Felici M, Klinger FG, Campolo F, Balistreri CR, Barchi M, Dolci S. To Be or Not to Be a Germ Cell: The Extragonadal Germ Cell Tumor Paradigm. Int J Mol Sci. 2021;22(11).
  280. Cattoni A, Albanese A. Case report: Fluctuating tumor markers in a boy with gonadotropin-releasing hormone-independent precocious puberty induced by a pineal germ cell tumor. Front Pediatr. 2022;10:940656.
  281. . !!! INVALID CITATION !!! (269, 271, 272).
  282. Yhoshu E, Lone YA, Mahajan JK, Singh UB. Hepatoblastoma with Precocious Puberty. J Indian Assoc Pediatr Surg. 2019;24(1):68-71.
  283. Hersmus R, van Bever Y, Wolffenbuttel KP, Biermann K, Cools M, Looijenga LH. The biology of germ cell tumors in disorders of sex development. Clin Genet. 2017;91(2):292-301.
  284. Shenker A, Laue L, Kosugi S, Merendino JJ, Jr., Minegishi T, Cutler GB, Jr. A constitutively activating mutation of the luteinizing hormone receptor in familial male precocious puberty. Nature. 1993;365(6447):652-4.
  285. Themmen APN, Huhtaniemi IT. Mutations of gonadotropins and gonadotropin receptors: elucidating the physiology and pathophysiology of pituitary-gonadal function. Endocr Rev. 2000;21(5):551-83.
  286. Gurnurkar S, DiLillo E, Carakushansky M. A Case of Familial Male-limited Precocious Puberty with a Novel Mutation. J Clin Res Pediatr Endocrinol. 2021;13(2):239-44.
  287. Haddad NG, Eugster EA. Peripheral precocious puberty including congenital adrenal hyperplasia: causes, consequences, management and outcomes. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101273.
  288. Cunha-Silva M, Brito VN, Macedo DB, Bessa DS, Ramos CO, Lima LG, et al. Spontaneous fertility in a male patient with testotoxicosis despite suppression of FSH levels. Hum Reprod. 2018;33(5):914-8.
  289. Kooij CD, Mavinkurve-Groothuis AMC, Kremer Hovinga ICL, Looijenga LHJ, Rinne T, Giltay JC, et al. Familial Male-limited Precocious Puberty (FMPP) and Testicular Germ Cell Tumors. J Clin Endocrinol Metab. 2022;107(11):3035-44.
  290. Reiter EO, Mauras N, McCormick K, Kulshreshtha B, Amrhein J, De Luca F, et al. Bicalutamide plus anastrozole for the treatment of gonadotropin-independent precocious puberty in boys with testotoxicosis: a phase II, open-label pilot study (BATT). J Pediatr Endocrinol Metab. 2010;23(10):999-1009.
  291. Leschek EW, Flor AC, Bryant JC, Jones JV, Barnes KM, Cutler GB, Jr. Effect of Antiandrogen, Aromatase Inhibitor, and Gonadotropin-releasing Hormone Analog on Adult Height in Familial Male Precocious Puberty. J Pediatr. 2017;190:229-35.
  292. Flippo C, Kolli V, Andrew M, Berger S, Bhatti T, Boyce AM, et al. Precocious Puberty in a Boy With Bilateral Leydig Cell Tumors due to a Somatic Gain-of-Function LHCGR Variant. J Endocr Soc. 2022;6(10):bvac127.
  293. Latronico AC, Lins TS, Brito VN, Arnhold IJ, Mendonca BB. The effect of distinct activating mutations of the luteinizing hormone receptor gene on the pituitary-gonadal axis in both sexes. Clin Endocrinol (Oxf). 2000;53(5):609-13.
  294. Vanwyk JJ, Grumbach MM. Syndrome of Precocious Menstruation and Galactorrhea in Juvenile Hypothyroidism - an Example of Hormonal Overlap in Pituitary Feedback. J Pediatr-Us. 1960;57(3):416-35.
  295. Kusuma Boddu S, Ayyavoo A, Hebbal Nagarajappa V, Kalenahalli KV, Muruda S, Palany R. Van Wyk Grumbach Syndrome and Ovarian Hyperstimulation in Juvenile Primary Hypothyroidism: Lessons From a 30-Case Cohort. J Endocr Soc. 2023;7(6):bvad042.
  296. Reddy P, Tiwari K, Kulkarni A, Parikh K, Khubchandani R. Van Wyk Grumbach Syndrome: A Rare Consequence of Hypothyroidism. Indian J Pediatr. 2018;85(11):1028-30.
  297. Zhang H, Geng N, Wang Y, Tian W, Xue F. Van Wyk and Grumbach syndrome: two case reports and review of the published work. J Obstet Gynaecol Res. 2014;40(2):607-10.
  298. Baranowski E, Hogler W. An unusual presentation of acquired hypothyroidism: the Van Wyk-Grumbach syndrome. Eur J Endocrinol. 2012;166(3):537-42.
  299. Anasti JN, Flack MR, Froehlich J, Nelson LM, Nisula BC. A potential novel mechanism for precocious puberty in juvenile hypothyroidism. J Clin Endocrinol Metab. 1995;80(1):276-9.
  300. Polat S, Kulle A, Karaca Z, Akkurt I, Kurtoglu S, Kelestimur F, et al. Characterisation of three novel CYP11B1 mutations in classic and non-classic 11beta-hydroxylase deficiency. Eur J Endocrinol. 2014;170(5):697-706.
  301. Armengaud JB, Charkaluk ML, Trivin C, Tardy V, Breart G, Brauner R, et al. Precocious pubarche: distinguishing late-onset congenital adrenal hyperplasia from premature adrenarche. J Clin Endocrinol Metab. 2009;94(8):2835-40.
  302. Miller WL. Congenital Adrenal Hyperplasia: Time to Replace 17OHP with 21-Deoxycortisol. Horm Res Paediatr. 2019;91(6):416-20.
  303. Sarafoglou K, Merke DP, Reisch N, Claahsen-van der Grinten H, Falhammar H, Auchus RJ. Interpretation of Steroid Biomarkers in 21-Hydroxylase Deficiency and Their Use in Disease Management. J Clin Endocrinol Metab. 2023;108(9):2154-75.
  304. Mallappa A, Merke DP. Management challenges and therapeutic advances in congenital adrenal hyperplasia. Nat Rev Endocrinol. 2022;18(6):337-52.
  305. Witchel SF. Congenital Adrenal Hyperplasia. J Pediatr Adolesc Gynecol. 2017;30(5):520-34.
  306. Wieneke JA, Thompson LD, Heffess CS. Adrenal cortical neoplasms in the pediatric population: a clinicopathologic and immunophenotypic analysis of 83 patients. Am J Surg Pathol. 2003;27(7):867-81.
  307. . !!! INVALID CITATION !!! (297).
  308. Custodio G, Komechen H, Figueiredo FR, Fachin ND, Pianovski MA, Figueiredo BC. Molecular epidemiology of adrenocortical tumors in southern Brazil. Mol Cell Endocrinol. 2012;351(1):44-51.
  309. Lapunzina P. Risk of tumorigenesis in overgrowth syndromes: a comprehensive review. Am J Med Genet C Semin Med Genet. 2005;137C(1):53-71.
  310. Clay MR, Pinto EM, Fishbein L, Else T, Kiseljak-Vassiliades K. Pathological and Genetic Stratification for Management of Adrenocortical Carcinoma. J Clin Endocrinol Metab. 2022;107(4):1159-69.
  311. Kaplowitz PB. For Premature Thelarche and Premature Adrenarche, the Case for Waiting before Testing. Horm Res Paediatr. 2020;93(9-10):573-6.
  312. Freedman SM, Kreitzer PM, Elkowitz SS, Soberman N, Leonidas JC. Ovarian microcysts in girls with isolated premature thelarche. J Pediatr. 1993;122(2):246-9.
  313. Witchel SF, Pinto B, Burghard AC, Oberfield SE. Update on adrenarche. Curr Opin Pediatr. 2020;32(4):574-81.
  314. Kaya G, Abali ZY, Bas F, Poyrazoglu S, Darendeliler F. Body mass index at the presentation of premature adrenarche is associated with components of metabolic syndrome at puberty. European Journal of Pediatrics. 2018;177(11):1593-601.
  315. Ibanez L, Ong K, de Zegher F, Marcos MV, del Rio L, Dunger DB. Fat distribution in non-obese girls with and without precocious pubarche: central adiposity related to insulinaemia and androgenaemia from prepuberty to postmenarche. Clin Endocrinol (Oxf). 2003;58(3):372-9.
  316. Ibanez L, Potau N, Virdis R, Zampolli M, Terzi C, Gussinye M, et al. Postpubertal outcome in girls diagnosed of premature pubarche during childhood: increased frequency of functional ovarian hyperandrogenism. J Clin Endocrinol Metab. 1993;76(6):1599-603.
  317. Tennila J, Jaaskelainen J, Utriainen P, Voutilainen R, Hakkinen M, Auriola S, et al. PCOS Features and Steroid Profiles Among Young Adult Women with a History of Premature Adrenarche. J Clin Endocrinol Metab. 2021;106(9):e3335-e45.
  318. Ramsey JT, Li Y, Arao Y, Naidu A, Coons LA, Diaz A, et al. Lavender Products Associated With Premature Thelarche and Prepubertal Gynecomastia: Case Reports and Endocrine-Disrupting Chemical Activities. J Clin Endocrinol Metab. 2019;104(11):5393-405.
  319. Kim SH, Park MJ. Effects of phytoestrogen on sexual development. Korean J Pediatr. 2012;55(8):265-71.
  320. Cleemann L, Holm K. [Precocious pseudopuberty in a seven year-old girl due to estrogen treatment of labial adhesion]. Ugeskr Laeger. 2011;173(20):1435-6.
  321. Guarneri MP, Brambilla G, Loizzo A, Colombo I, Chiumello G. Estrogen exposure in a child from hair lotion used by her mother: clinical and hair analysis data. Clin Toxicol (Phila). 2008;46(8):762-4.
  322. Martinez-Pajares JD, Diaz-Morales O, Ramos-Diaz JC, Gomez-Fernandez E. Peripheral precocious puberty due to inadvertent exposure to testosterone: case report and review of the literature. J Pediatr Endocrinol Metab. 2012;25(9-10):1007-12.
  323. Castiello F, Freire C. Exposure to non-persistent pesticides and puberty timing: a systematic review of the epidemiological evidence. Eur J Endocrinol. 2021;184(6):733-49.
  324. Watkins DJ, Sanchez BN, Tellez-Rojo MM, Lee JM, Mercado-Garcia A, Blank-Goldenberg C, et al. Phthalate and bisphenol A exposure during in utero windows of susceptibility in relation to reproductive hormones and pubertal development in girls. Environ Res. 2017;159:143-51.
  325. Lee JE, Jung HW, Lee YJ, Lee YA. Early-life exposure to endocrine-disrupting chemicals and pubertal development in girls. Ann Pediatr Endocrinol Metab. 2019;24(2):78-91.
  326. Franssen D, Svingen T, Lopez Rodriguez D, Van Duursen M, Boberg J, Parent AS. A Putative Adverse Outcome Pathway Network for Disrupted Female Pubertal Onset to Improve Testing and Regulation of Endocrine Disrupting Chemicals. Neuroendocrinology. 2022;112(2):101-14.
  327. Taylor KW, Howdeshell KL, Bommarito PA, Sibrizzi CA, Blain RB, Magnuson K, et al. Systematic evidence mapping informs a class-based approach to assessing personal care products and pubertal timing. Environ Int. 2023;181:108307.
  328. Ahn C, Jeung EB. Endocrine-Disrupting Chemicals and Disease Endpoints. Int J Mol Sci. 2023;24(6).
  329. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev. 2015;36(6):E1-E150.
  330. Uldbjerg CS, Koch T, Lim YH, Gregersen LS, Olesen CS, Andersson AM, et al. Prenatal and postnatal exposures to endocrine disrupting chemicals and timing of pubertal onset in girls and boys: a systematic review and meta-analysis. Hum Reprod Update. 2022;28(5):687-716.
  331. De Sanctis V, Rigon F, Bernasconi S, Bianchin L, Bona G, Bozzola M, et al. Age at Menarche and Menstrual Abnormalities in Adolescence: Does it Matter? The Evidence from a Large Survey among Italian Secondary Schoolgirls. Indian J Pediatr. 2019;86(Suppl 1):34-41.
  332. Reindollar RH, Byrd JR, McDonough PG. Delayed sexual development: a study of 252 patients. Am J Obstet Gynecol. 1981;140(4):371-80.
  333. Herman-Giddens ME, Steffes J, Harris D, Slora E, Hussey M, Dowshen SA, et al. Secondary sexual characteristics in boys: data from the Pediatric Research in Office Settings Network. Pediatrics. 2012;130(5):e1058-68.
  334. Jonsdottir-Lewis E, Feld A, Ciarlo R, Denhoff E, Feldman HA, Chan YM. Timing of Pubertal Onset in Girls and Boys With Constitutional Delay. J Clin Endocrinol Metab. 2021;106(9):e3693-e703.
  335. Barroso PS, Jorge AAL, Lerario AM, Montenegro LR, Vasques GA, Lima Amato LG, et al. Clinical and Genetic Characterization of a Constitutional Delay of Growth and Puberty Cohort. Neuroendocrinology. 2020;110(11-12):959-66.
  336. Sukumar SP, Bhansali A, Sachdeva N, Ahuja CK, Gorsi U, Jarial KD, et al. Diagnostic utility of testosterone priming prior to dynamic tests to differentiate constitutional delay in puberty from isolated hypogonadotropic hypogonadism. Clin Endocrinol (Oxf). 2017;86(5):717-24.
  337. Galazzi E, Improda N, Cerbone M, Soranna D, Moro M, Fatti LM, et al. Clinical benefits of sex steroids given as a priming prior to GH provocative test or as a growth-promoting therapy in peripubertal growth delays: Results of a retrospective study among ENDO-ERN centres. Clin Endocrinol (Oxf). 2021;94(2):219-28.
  338. Vezzoli V, Hrvat F, Goggi G, Federici S, Cangiano B, Quinton R, et al. Genetic architecture of self-limited delayed puberty and congenital hypogonadotropic hypogonadism. Front Endocrinol. 2023;13.
  339. Aung Y, Kokotsis V, Yin KN, Banerjee K, Butler G, Dattani MT, et al. Key features of puberty onset and progression can help distinguish self-limited delayed puberty from congenital hypogonadotrophic hypogonadism. Front Endocrinol (Lausanne). 2023;14:1226839.
  340. Gohil A, Eugster EA. Delayed and Precocious Puberty: Genetic Underpinnings and Treatments. Endocrin Metab Clin. 2020;49(4):741-+.
  341. Cassatella D, Howard SR, Acierno JS, Xu C, Papadakis GE, Santoni FA, et al. Congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty have distinct genetic architectures. European Journal of Endocrinology. 2018;178(4):377-88.
  342. Zhu J, Choa RE, Guo MH, Plummer L, Buck C, Palmert MR, et al. A shared genetic basis for self-limited delayed puberty and idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2015;100(4):E646-54.
  343. Cui X, Cui Y, Shi L, Luan J, Zhou X, Han J. A basic understanding of Turner syndrome: Incidence, complications, diagnosis, and treatment. Intractable Rare Dis Res. 2018;7(4):223-8.
  344. Cameron-Pimblett A, La Rosa C, King TFJ, Davies MC, Conway GS. The Turner syndrome life course project: Karyotype-phenotype analyses across the lifespan. Clin Endocrinol (Oxf). 2017;87(5):532-8.
  345. Gravholt CH, Andersen NH, Conway GS, Dekkers OM, Geffner ME, Klein KO, et al. Clinical practice guidelines for the care of girls and women with Turner syndrome: proceedings from the 2016 Cincinnati International Turner Syndrome Meeting. Eur J Endocrinol. 2017;177(3):G1-G70.
  346. Chan YM, Lippincott MF, Sales Barroso P, Alleyn C, Brodsky J, Granados H, et al. Using Kisspeptin to Predict Pubertal Outcomes for Youth With Pubertal Delay. J Clin Endocrinol Metab. 2020;105(8):e2717-25.
  347. Bojesen A, Juul S, Gravholt CH. Prenatal and postnatal prevalence of Klinefelter syndrome: a national registry study. J Clin Endocrinol Metab. 2003;88(2):622-6.
  348. Tian SY, Li Y, Zhao LM, He HY. [Clinicopathological characteristics of Klinefelter syndrome: a testicular biopsy analysis of 87 cases]. Zhonghua Bing Li Xue Za Zhi. 2023;52(4):341-6.
  349. Hamzik MP, Gropman AL, Brooks MR, Powell S, Sadeghin T, Samango-Sprouse CA. The Effect of Hormonal Therapy on the Behavioral Outcomes in 47,XXY (Klinefelter Syndrome) between 7 and 12 Years of Age. Genes (Basel). 2023;14(7).
  350. Lazos-Ochoa M, Aguirre D, Nieto K, Pena R, Palma I, Kofman S, et al. Extragonadal mediastinal germ cell tumors are often associated with Klinefelter syndrome. Modern Pathol. 2006;19:137-.
  351. Lopez Krabbe HV, Holm Petersen J, Asserhoj LL, Johannsen TH, Christiansen P, Jensen RB, et al. Reproductive hormones, bone mineral content, body composition, and testosterone therapy in boys and adolescents with Klinefelter syndrome. Endocr Connect. 2023;12(7).
  352. Rohayem J, Nieschlag E, Zitzmann M, Kliesch S. Testicular function during puberty and young adulthood in patients with Klinefelter's syndrome with and without spermatozoa in seminal fluid. Andrology. 2016;4(6):1178-86.
  353. Masterson TA, 3rd, Nassau DE, Ramasamy R. A clinical algorithm for management of fertility in adolescents with the Klinefelter syndrome. Curr Opin Urol. 2020;30(3):324-7.
  354. Vena W, Carrone F, Delbarba A, Akpojiyovbi O, Pezzaioli LC, Facondo P, et al. Body composition, trabecular bone score and vertebral fractures in subjects with Klinefelter syndrome. J Endocrinol Invest. 2023;46(2):297-304.
  355. Butler G, Srirangalingam U, Faithfull J, Sangster P, Senniappan S, Mitchell R. Klinefelter syndrome: going beyond the diagnosis. Arch Dis Child. 2023;108(3):166-71.
  356. Tanner M, Miettinen PJ, Hero M, Toppari J, Raivio T. Onset and progression of puberty in Klinefelter syndrome. Clin Endocrinol (Oxf). 2022;96(3):363-70.
  357. Cools M, Nordenstrom A, Robeva R, Hall J, Westerveld P, Fluck C, et al. Caring for individuals with a difference of sex development (DSD): a Consensus Statement. Nat Rev Endocrinol. 2018;14(7):415-29.
  358. Rosario R, Anderson R. The molecular mechanisms that underlie fragile X-associated premature ovarian insufficiency: is it RNA or protein based? Mol Hum Reprod. 2020;26(10):727-37.
  359. Biancalana V, Glaeser D, McQuaid S, Steinbach P. EMQN best practice guidelines for the molecular genetic testing and reporting of fragile X syndrome and other fragile X-associated disorders. Eur J Hum Genet. 2015;23(4):417-25.
  360. Yatsenko SA, Rajkovic A. Genetics of human female infertilitydagger. Biol Reprod. 2019;101(3):549-66.
  361. Flechtner I, Viaud M, Kariyawasam D, Perrissin-Fabert M, Bidet M, Bachelot A, et al. Puberty and fertility in classic galactosemia. Endocr Connect. 2021;10(2):240-7.
  362. Gubbels CS, Land JA, Rubio-Gozalbo ME. Fertility and impact of pregnancies on the mother and child in classic galactosemia. Obstet Gynecol Surv. 2008;63(5):334-43.
  363. Frederick AB, Zinsli AM, Carlock G, Conneely K, Fridovich-Keil JL. Presentation, progression, and predictors of ovarian insufficiency in classic galactosemia. J Inherit Metab Dis. 2018;41(5):785-90.
  364. Derks B, Rivera-Cruz G, Hagen-Lillevik S, Vos EN, Demirbas D, Lai K, et al. The hypergonadotropic hypogonadism conundrum of classic galactosemia. Hum Reprod Update. 2023;29(2):246-58.
  365. Rubio-Gozalbo ME, Gubbels CS, Bakker JA, Menheere PP, Wodzig WK, Land JA. Gonadal function in male and female patients with classic galactosemia. Hum Reprod Update. 2010;16(2):177-88.
  366. Schweitzer S, Shin Y, Jakobs C, Brodehl J. Long-term outcome in 134 patients with galactosaemia. Eur J Pediatr. 1993;152(1):36-43.
  367. Waggoner DD, Buist NR, Donnell GN. Long-term prognosis in galactosaemia: results of a survey of 350 cases. J Inherit Metab Dis. 1990;13(6):802-18.
  368. Waldstreicher J, Seminara SB, Jameson JL, Geyer A, Nachtigall LB, Boepple PA, et al. The genetic and clinical heterogeneity of gonadotropin-releasing hormone deficiency in the human. J Clin Endocrinol Metab. 1996;81(12):4388-95.
  369. Sykiotis GP, Pitteloud N, Seminara SB, Kaiser UB, Crowley WF, Jr. Deciphering genetic disease in the genomic era: the model of GnRH deficiency. Sci Transl Med. 2010;2(32):32rv2.
  370. Sykiotis GP, Plummer L, Hughes VA, Au M, Durrani S, Nayak-Young S, et al. Oligogenic basis of isolated gonadotropin-releasing hormone deficiency. Proc Natl Acad Sci U S A. 2010;107(34):15140-4.
  371. Linscott ML, Chung WCJ. Epigenomic control of gonadotrophin-releasing hormone neurone development and hypogonadotrophic hypogonadism. J Neuroendocrinol. 2020;32(6):e12860.
  372. Harrington J, Palmert MR. An Approach to the Patient With Delayed Puberty. J Clin Endocrinol Metab. 2022;107(6):1739-50.
  373. Molsted K, Kjaer I, Giwercman A, Vesterhauge S, Skakkebaek NE. Craniofacial morphology in patients with Kallmann's syndrome with and without cleft lip and palate. Cleft Palate Craniofac J. 1997;34(5):417-24.
  374. Cassatella D, Howard SR, Acierno JS, Xu C, Papadakis GE, Santoni FA, et al. Congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty have distinct genetic architectures. Eur J Endocrinol. 2018;178(4):377-88.
  375. Husebye ES, Anderson MS, Kampe O. Autoimmune Polyendocrine Syndromes. N Engl J Med. 2018;378(12):1132-41.
  376. McElreavey K, Jorgensen A, Eozenou C, Merel T, Bignon-Topalovic J, Tan DS, et al. Pathogenic variants in the DEAH-box RNA helicase DHX37 are a frequent cause of 46,XY gonadal dysgenesis and 46,XY testicular regression syndrome. Genet Med. 2020;22(1):150-9.
  377. Foster KL, Lee DJ, Witchel SF, Gordon CM. Ovarian Insufficiency and Fertility Preservation During and After Childhood Cancer Treatment. J Adolesc Young Adult Oncol. 2024.
  378. Matalliotakis M, Koliarakis I, Matalliotaki C, Trivli A, Hatzidaki E. Clinical manifestations, evaluation and management of hyperprolactinemia in adolescent and young girls: a brief review. Acta Biomed. 2019;90(1):149-57.
  379. Lee SL, Lim A, Munns C, Simm PJ, Zacharin M. Effect of Testosterone Treatment for Delayed Puberty in Duchenne Muscular Dystrophy. Horm Res Paediatr. 2020;93(2):108-18.
  380. Ward LM, Weber DR. Growth, pubertal development, and skeletal health in boys with Duchenne Muscular Dystrophy. Curr Opin Endocrinol Diabetes Obes. 2019;26(1):39-48.
  381. Esquivel-Zuniga R, Rogol AD. Functional hypogonadism in adolescence: an overlooked cause of secondary hypogonadism. Endocr Connect. 2023;12(11).
  382. Thavaraputta S, Ungprasert P, Witchel SF, Fazeli PK. Anorexia nervosa and adrenal hormones: a systematic review and meta-analysis. Eur J Endocrinol. 2023;189(3):S64-S73.
  383. Misra M, Golden NH, Katzman DK. State of the art systematic review of bone disease in anorexia nervosa. Int J Eat Disord. 2016;49(3):276-92.
  384. Odle AK, Akhter N, Syed MM, Allensworth-James ML, Benes H, Melgar Castillo AI, et al. Leptin Regulation of Gonadotrope Gonadotropin-Releasing Hormone Receptors As a Metabolic Checkpoint and Gateway to Reproductive Competence. Front Endocrinol (Lausanne). 2017;8:367.
  385. Vazquez MJ, Velasco I, Tena-Sempere M. Novel mechanisms for the metabolic control of puberty: implications for pubertal alterations in early-onset obesity and malnutrition. J Endocrinol. 2019;242(2):R51-R65.
  386. Cano Sokoloff N, Misra M, Ackerman KE. Exercise, Training, and the Hypothalamic-Pituitary-Gonadal Axis in Men and Women. Front Horm Res. 2016;47:27-43.
  387. Gordon CM, Ackerman KE, Berga SL, Kaplan JR, Mastorakos G, Misra M, et al. Functional Hypothalamic Amenorrhea: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017;102(5):1413-39.
  388. De Lorenzo A, Noce A, Moriconi E, Rampello T, Marrone G, Di Daniele N, et al. MOSH Syndrome (Male Obesity Secondary Hypogonadism): Clinical Assessment and Possible Therapeutic Approaches. Nutrients. 2018;10(4).
  389. Federici S, Goggi G, Quinton R, Giovanelli L, Persani L, Cangiano B, et al. New and Consolidated Therapeutic Options for Pubertal Induction in Hypogonadism: In-depth Review of the Literature. Endocr Rev. 2022;43(5):824-51.
  390. Szeliga A, Podfigurna A, Bala G, Meczekalski B. Kisspeptin and neurokinin B analogs use in gynecological endocrinology: where do we stand? J Endocrinol Invest. 2020;43(5):555-61.
  391. Alexander EC, Faruqi D, Farquhar R, Unadkat A, Ng Yin K, Hoskyns R, et al. Gonadotropins for pubertal induction in males with hypogonadotropic hypogonadism: systematic review and meta-analysis. Eur J Endocrinol. 2024;190(1):S1-S11.
  392. Stancampiano MR, Lucas-Herald AK, Russo G, Rogol AD, Ahmed SF. Testosterone Therapy in Adolescent Boys: The Need for a Structured Approach. Horm Res Paediatr. 2019;92(4):215-28.
  393. Raivio T, Falardeau J, Dwyer A, Quinton R, Hayes FJ, Hughes VA, et al. Reversal of idiopathic hypogonadotropic hypogonadism. N Engl J Med. 2007;357(9):863-73.
  394. Sun T, Xu W, Chen Y, Niu Y, Wang T, Wang S, et al. Reversal of idiopathic hypogonadotropic hypogonadism in a Chinese male cohort. Andrologia. 2022;54(11):e14583.
  395. Klein KO, Phillips SA. Review of Hormone Replacement Therapy in Girls and Adolescents with Hypogonadism. J Pediatr Adolesc Gynecol. 2019;32(5):460-8.
  396. Cheuiche AV, da Silveira LG, de Paula LCP, Lucena IRS, Silveiro SP. Diagnosis and management of precocious sexual maturation: an updated review. Eur J Pediatr. 2021;180(10):3073-87.
  397. Ibanez L, Ferrer A, Marcos MV, Hierro FR, de Zegher F. Early puberty: rapid progression and reduced final height in girls with low birth weight. Pediatrics. 2000;106(5):E72.
  398. Young J, Xu C, Papadakis GE, Acierno JS, Maione L, Hietamaki J, et al. Clinical Management of Congenital Hypogonadotropic Hypogonadism. Endocr Rev. 2019;40(2):669-710.
  399. Varimo T, Hero M, Laitinen EM, Miettinen PJ, Tommiska J, Kansakoski J, et al. Childhood growth in boys with congenital hypogonadotropic hypogonadism. Pediatr Res. 2016;79(5):705-9.
  400. Varimo T, Miettinen PJ, Kansakoski J, Raivio T, Hero M. Congenital hypogonadotropic hypogonadism, functional hypogonadotropism or constitutional delay of growth and puberty? An analysis of a large patient series from a single tertiary center. Hum Reprod. 2017;32(1):147-53.
  401. Neely EK, Hintz RL, Wilson DM, Lee PA, Gautier T, Argente J, et al. Normal ranges for immunochemiluminometric gonadotropin assays. J Pediatr. 1995;127(1):40-6.
  402. Martinez-Aguayo A, Hernández MI, Capurro T, Peña V, Avila A, Salazar T, et al. Leuprolide acetate gonadotrophin response patterns during female puberty. Clin Endocrinol (Oxf). 2010;72(4):489-95.
  403. Cao R, Liu J, Fu P, Zhou Y, Li Z, Liu P. The Diagnostic Utility of the Basal Luteinizing Hormone Level and Single 60-Minute Post GnRH Agonist Stimulation Test for Idiopathic Central Precocious Puberty in Girls. Front Endocrinol (Lausanne). 2021;12:713880.
  404. Howard SR. Interpretation of reproductive hormones before, during and after the pubertal transition-Identifying health and disordered puberty. Clin Endocrinol (Oxf). 2021;95(5):702-15.
  405. Bizzarri C, Spadoni GL, Bottaro G, Montanari G, Giannone G, Cappa M, et al. The response to gonadotropin releasing hormone (GnRH) stimulation test does not predict the progression to true precocious puberty in girls with onset of premature thelarche in the first three years of life. J Clin Endocrinol Metab. 2014;99(2):433-9.
  406. Vestergaard ET, Schjorring ME, Kamperis K, Petersen KK, Rittig S, Juul A, et al. The follicle-stimulating hormone (FSH) and luteinizing hormone (LH) response to a gonadotropin-releasing hormone analogue test in healthy prepubertal girls aged 10 months to 6 years. Eur J Endocrinol. 2017;176(6):747-53.
  407. Harrington J, Palmert MR. Clinical review: Distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism: critical appraisal of available diagnostic tests. J Clin Endocrinol Metab. 2012;97(9):3056-67.
  408. Degros V, Cortet-Rudelli C, Soudan B, Dewailly D. The human chorionic gonadotropin test is more powerful than the gonadotropin-releasing hormone agonist test to discriminate male isolated hypogonadotropic hypogonadism from constitutional delayed puberty. European Journal of Endocrinology. 2003;149(1):23-9.
  409. Chaudhary S, Walia R, Bhansali A, Dayal D, Sachdeva N, Singh T, et al. FSH-stimulated Inhibin B (FSH-iB): A Novel Marker for the Accurate Prediction of Pubertal Outcome in Delayed Puberty. J Clin Endocrinol Metab. 2021;106(9):e3495-e505.
  410. Turcu AF, Mallappa A, Nella AA, Chen X, Zhao L, Nanba AT, et al. 24-Hour Profiles of 11-Oxygenated C(19) Steroids and Delta(5)-Steroid Sulfates during Oral and Continuous Subcutaneous Glucocorticoids in 21-Hydroxylase Deficiency. Front Endocrinol (Lausanne). 2021;12:751191.
  411. Trost LW, Mulhall JP. Challenges in Testosterone Measurement, Data Interpretation, and Methodological Appraisal of Interventional Trials. J Sex Med. 2016;13(7):1029-46.
  412. French D. Clinical utility of laboratory developed mass spectrometry assays for steroid hormone testing. J Mass Spectrom Adv Clin Lab. 2023;28:13-9.
  413. Chin VL, Cai Z, Lam L, Shah B, Zhou P. Evaluation of puberty by verifying spontaneous and stimulated gonadotropin values in girls. J Pediatr Endocrinol Metab. 2015;28(3-4):387-92.
  414. Abbara A, Eng PC, Phylactou M, Clarke SA, Mills E, Chia G, et al. Kisspeptin-54 Accurately Identifies Hypothalamic Gonadotropin-Releasing Hormone Neuronal Dysfunction in Men with Congenital Hypogonadotropic Hypogonadism. Neuroendocrinology. 2021;111(12):1176-86.
  415. Rosenfield RL. Normal and Premature Adrenarche. Endocr Rev. 2021;42(6):783-814.
  416. Chesover AD, Millar H, Sepiashvili L, Adeli K, Palmert MR, Hamilton J. Screening for Nonclassic Congenital Adrenal Hyperplasia in the Era of Liquid Chromatography-Tandem Mass Spectrometry. J Endocr Soc. 2020;4(2):bvz030.
  417. Gao Y, Du Q, Liu L, Liao Z. Serum inhibin B for differentiating between congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty: a systematic review and meta-analysis. Endocrine. 2021;72(3):633-43.
  418. Binder G, Schweizer R, Blumenstock G, Braun R. Inhibin B plus LH vs GnRH agonist test for distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism in boys. Clin Endocrinol (Oxf). 2015;82(1):100-5.
  419. Mosbah H, Bouvattier C, Maione L, Trabado S, De Filippo G, Cartes A, et al. GnRH stimulation testing and serum inhibin B in males: insufficient specificity for discriminating between congenital hypogonadotropic hypogonadism from constitutional delay of growth and puberty. Hum Reprod. 2020;35(10):2312-22.
  420. Albrethsen J, Ljubicic ML, Juul A. Longitudinal Increases in Serum Insulin-like Factor 3 and Testosterone Determined by LC-MS/MS in Pubertal Danish Boys. J Clin Endocrinol Metab. 2020;105(10).
  421. Segal TY, Mehta A, Anazodo A, Hindmarsh PC, Dattani MT. Role of gonadotropin-releasing hormone and human chorionic gonadotropin stimulation tests in differentiating patients with hypogonadotropic hypogonadism from those with constitutional delay of growth and puberty. J Clin Endocrinol Metab. 2009;94(3):780-5.
  422. Greulich WW PS. Radiographic Atlas of Skeletal Development of the Hand and Wrist. 2nd ed: Stanford University Press; 1999.
  423. Tanner JM WR, Cameron N, et al.,. Assessment of Skeletal Maturity and Prediction of Adult Height (TW2 Method). 2nd ed: Goldstein H (Ed), Academic Press; 1983.
  424. Klein KO, Newfield RS, Hassink SG. Bone maturation along the spectrum from normal weight to obesity: a complex interplay of sex, growth factors and weight gain. J Pediatr Endocrinol Metab. 2016;29(3):311-8.
  425. Oh MS, Kim S, Lee J, Lee MS, Kim YJ, Kang KS. Factors associated with Advanced Bone Age in Overweight and Obese Children. Pediatr Gastroentero. 2020;23(1):89-97.
  426. Bar A, Linder B, Sobel EH, Saenger P, Dimartinonardi J. Bayley-Pinneau Method of Height Prediction in Girls with Central Precocious Puberty - Correlation with Adult Height. J Pediatr-Us. 1995;126(6):955-8.
  427. Eitel KB, Eugster EA. Differences in Bone Age Readings between Pediatric Endocrinologists and Radiologists. Endocrine Practice. 2020;26(3):328-31.
  428. Wang X, Zhou B, Gong P, Zhang T, Mo Y, Tang J, et al. Artificial Intelligence-Assisted Bone Age Assessment to Improve the Accuracy and Consistency of Physicians With Different Levels of Experience. Frontiers in Pediatrics. 2022;10.
  429. Thodberg HH, Thodberg B, Ahlkvist J, Offiah AC. Autonomous artificial intelligence in pediatric radiology: the use and perception of BoneXpert for bone age assessment. Pediatr Radiol. 2022;52(7):1338-46.
  430. de Vries L, Horev G, Schwartz M, Phillip M. Ultrasonographic and clinical parameters for early differentiation between precocious puberty and premature thelarche. Eur J Endocrinol. 2006;154(6):891-8.
  431. Sathasivam A, Rosenberg HK, Shapiro S, Wang H, Rapaport R. Pelvic ultrasonography in the evaluation of central precocious puberty: comparison with leuprolide stimulation test. J Pediatr. 2011;159(3):490-5.
  432. Lee SH, Joo EY, Lee JE, Jun YH, Kim MY. The Diagnostic Value of Pelvic Ultrasound in Girls with Central Precocious Puberty. Chonnam Med J. 2016;52(1):70-4.
  433. Battaglia C, Mancini F, Regnani G, Persico N, Iughetti L, De Aloysio D. Pelvic ultrasound and color Doppler findings in different isosexual precocities. Ultrasound Obstet Gynecol. 2003;22(3):277-83.
  434. Long MG, Boultbee JE, Hanson ME, Begent RH. Doppler time velocity waveform studies of the uterine artery and uterus. Br J Obstet Gynaecol. 1989;96(5):588-93.
  435. Paesano PL, Colantoni C, Mora S, di Lascio A, Ferrario M, Esposito A, et al. Validation of an Accurate and Noninvasive Tool to Exclude Female Precocious Puberty: Pelvic Ultrasound With Uterine Artery Pulsatility Index. Am J Roentgenol. 2019;213(2):451-7.
  436. Cheuiche AV, Moro C, Lucena IRS, de Paula LCP, Silveiro SP. Accuracy of doppler assessment of the uterine arteries for the diagnosis of pubertal onset in girls: a scoping review. Sci Rep. 2023;13(1):5791.
  437. Kafi SE, Alagha E, Shazly MA, Al-Agha A. Pseudo-precocious Puberty Associated with an Adrenocortical Tumor in a Young Child. Cureus. 2019;11(12).
  438. Alagha E, Kafi SE, Shazly MA, Al-Agha A. Precocious Puberty Associated with Testicular Hormone-secreting Leydig Cell Tumor. Cureus. 2019;11(12):e6441.
  439. Eyer de Jesus L, Paz de Oliveira AP, Porto LC, Dekermacher S. Testicular adrenal rest tumors - Epidemiology, diagnosis and treatment. J Pediatr Urol. 2023.
  440. Cantas-Orsdemir S, Garb JL, Allen HF. Prevalence of cranial MRI findings in girls with central precocious puberty: a systematic review and meta-analysis. J Pediatr Endocrinol Metab. 2018;31(7):701-10.
  441. Pedicelli S, Alessio P, Scire G, Cappa M, Cianfarani S. Routine screening by brain magnetic resonance imaging is not indicated in every girl with onset of puberty between the ages of 6 and 8 years. J Clin Endocrinol Metab. 2014;99(12):4455-61.
  442. Kaplowitz PB. Do 6-8 year old girls with central precocious puberty need routine brain imaging? Int J Pediatr Endocrinol. 2016;2016:9.
  443. Mogensen SS, Aksglaede L, Mouritsen A, Sorensen K, Main KM, Gideon P, et al. Pathological and Incidental Findings on Brain MRI in a Single-Center Study of 229 Consecutive Girls with Early or Precocious Puberty. Plos One. 2012;7(1).
  444. Hansen AB, Renault CH, Wojdemann D, Gideon P, Juul A, Jensen RB. Neuroimaging in 205 consecutive Children Diagnosed with Central Precocious Puberty in Denmark. Pediatr Res. 2023;93(1):125-30.
  445. Kim SH, Ahn MB, Cho WK, Cho KS, Jung MH, Suh BK. Findings of Brain Magnetic Resonance Imaging in Girls with Central Precocious Puberty Compared with Girls with Chronic or Recurrent Headache. J Clin Med. 2021;10(10).
  446. Eugster EA. Update on Precocious Puberty in Girls. J Pediatr Adol Gynec. 2019;32(5):455-9.
  447. Yoon JS, So CH, Lee HS, Lim JS, Hwang JS. The prevalence of brain abnormalities in boys with central precocious puberty may be overestimated. Plos One. 2018;13(4).
  448. Kang JY, Kim SH, Kim H, Ki H, Lee MH. Pituitary magnetic resonance imaging abnormalities in young female patients with hypogonadotropic hypogonadism. Obstet Gynecol Sci. 2019;62(4):249-57.
  449. Hacquart T, Ltaief-Boudrigua A, Jeannerod C, Hannoun S, Raverot G, Pugeat M, et al. Reconsidering olfactory bulb magnetic resonance patterns in Kallmann syndrome. Ann Endocrinol (Paris). 2017;78(5):455-61.
  450. Sarfati J, Saveanu A, Young J. Pituitary stalk interruption and olfactory bulbs aplasia/hypoplasia in a man with Kallmann syndrome and reversible gonadotrope and somatotrope deficiencies. Endocrine. 2015;49(3):865-6.
  451. Elnaw EAA, Ibrahim AAB, Abdullah MA. Feminizing adrenocortical adenoma in a girl from a resource-limited setting: a case report. J Med Case Rep. 2021;15(1).
  452. Bulut S, Catli G, Filibeli BE, Manyas H, Ayranci I, Meral R, et al. Virilizing Adrenocortical Carcinoma Oncocytic Variant in a Child with Heterosexual Precocious Puberty and a Literature Review. J Pediatr Res. 2022;9(3):307-13.
  453. Ko JH, Lee HS, Hong J, Hwang JS. Virilizing adrenocortical carcinoma in a child with Turner syndrome and somatic TP53 gene mutation. European Journal of Pediatrics. 2010;169(4):501-4.
  454. Elnaw EAA, Ibrahim AAB, Abdullah MA. Feminizing adrenocortical adenoma in a girl from a resource-limited setting: a case report. J Med Case Rep. 2021;15(1):605.
  455. Gravholt CH, Viuff M, Just J, Sandahl K, Brun S, van der Velden J, et al. The Changing Face of Turner Syndrome. Endocr Rev. 2023;44(1):33-69.
  456. Correa Brito L, Rey RA. Taming Idiopathic Central Precocious Puberty: High Frequency of Imprinting Disorders in Familial Forms. J Clin Endocrinol Metab. 2023;108(8):e636-e7.
  457. Aguirre RS, Eugster EA. Central precocious puberty: From genetics to treatment. Best Pract Res Cl En. 2018;32(4):343-54.
  458. Howard SR, Dunkel L. Delayed Puberty-Phenotypic Diversity, Molecular Genetic Mechanisms, and Recent Discoveries. Endocr Rev. 2019;40(5):1285-317.
  459. Howard SR. Genes underlying delayed puberty. Mol Cell Endocrinol. 2018;476:119-28.
  460. Villanueva C, Jacobson-Dickman E, Xu C, Manouvrier S, Dwyer AA, Sykiotis GP, et al. Congenital hypogonadotropic hypogonadism with split hand/foot malformation: a clinical entity with a high frequency of FGFR1 mutations. Genet Med. 2015;17(8):651-9.
  461. LATRONICO A. Deciphering the genetic basis of central precocious puberty. Rev Esp Endocrinol Pediatr. 2023;14 Suppl(2):17-20
  462. Stecchini MF, Macedo DB, Reis AC, Abreu AP, Moreira AC, Castro M, et al. Time Course of Central Precocious Puberty Development Caused by an MKRN3 Gene Mutation: A Prismatic Case. Horm Res Paediatr. 2016;86(2):126-30.
  463. Teles MG, Bianco SDC, Brito VN, Trarbach EB, Kuohung W, Xu SY, et al. A GPR54-activating mutation in a patient with central precocious puberty. New Engl J Med. 2008;358(7):709-15.
  464. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Shagoury JK, et al. The GPR54 gene as a regulator of puberty. New Engl J Med. 2003;349(17):1614-U8.
  465. Krstevska-Konstantinova M, Jovanovska J, Tasic VB, Montenegro LR, Beneduzzi D, Silveira LF, et al. Mutational analysis of KISS1 and KISS1R in idiopathic central precocious puberty. J Pediatr Endocrinol Metab. 2014;27(1-2):199-201.
  466. Grandone A, Capristo C, Cirillo G, Sasso M, Umano GR, Mariani M, et al. Molecular Screening of MKRN3, DLK1, and KCNK9 Genes in Girls with Idiopathic Central Precocious Puberty. Horm Res Paediatr. 2017;88(3-4):194-200.
  467. Macedo DB, Kaiser UB. DLK1, Notch Signaling and the Timing of Puberty. Semin Reprod Med. 2019;37(4):174-81.
  468. Montenegro L, Labarta JI, Piovesan M, Canton APM, Corripio R, Soriano-Guillen L, et al. Novel Genetic and Biochemical Findings of DLK1 in Children with Central Precocious Puberty: A Brazilian-Spanish Study. J Clin Endocrinol Metab. 2020;105(10).
  469. Soares JM, de Holanda FS, Matsuzaki CN, Sorpreso ICE, Veiga ECD, de Abreu LC, et al. Analysis of the PvuII and XbaI polymorphisms in the estrogen receptor alpha gene in girls with central precocious puberty: a pilot study. Bmc Med Genet. 2018;19.
  470. Lee HS, Park HK, Kim KH, Ko JH, Kim YJ, Yi KH, et al. Estrogen receptor alpha gene analysis in girls with central precocious puberty. J Pediatr Endocr Met. 2013;26(7-8):645-9.
  471. Lee HS, Kim KH, Hwang JS. Association of aromatase (TTTA)n repeat polymorphisms with central precocious puberty in girls. Clin Endocrinol (Oxf). 2014;81(3):395-400.
  472. Tsuji-Hosokawa A, Matsuda N, Kurosawa K, Kashimada K, Morio T. A Case of MECP2 Duplication Syndrome with Gonadotropin-Dependent Precocious Puberty. Horm Res Paediatr. 2017;87(4):271-6.
  473. Canton APM, Krepischi ACV, Montenegro LR, Costa S, Rosenberg C, Steunou V, et al. Insights from the genetic characterization of central precocious puberty associated with multiple anomalies. Hum Reprod. 2021;36(2):506-18.
  474. Czako M, Till A, Zima J, Zsigmond A, Szabo A, Maasz A, et al. Xp11.2 Duplication in Females: Unique Features of a Rare Copy Number Variation. Front Genet. 2021;12:635458.
  475. Ludwig NG, Radaeli RF, da Silva MMX, Romero CM, Carrilho AJF, Bessa D, et al. A boy with Prader-Willi syndrome: unmasking precocious puberty during growth hormone replacement therapy. Arch Endocrin Metab. 2016;60(6):596-600.
  476. Hoffmann K, Heller R. Uniparental disomies 7 and 14. Best Pract Res Clin Endocrinol Metab. 2011;25(1):77-100.
  477. Partsch CJ, Japing I, Siebert R, Gosch A, Wessel A, Sippell WG, et al. Central precocious puberty in girls with Williams syndrome. J Pediatr. 2002;141(3):441-4.
  478. Utine GE, Alikasifoglu A, Alikasifoglu M, Tuncbilek E. Central precocious puberty in a girl with Williams syndrome: the result of treatment with GnRH analogue. Eur J Med Genet. 2006;49(1):79-82.
  479. Kuroki Y, Katsumata N, Eguchi T, Fukushima Y, Suwa S, Kajii T. Precocious puberty in Kabuki makeup syndrome. J Pediatr. 1987;110(5):750-2.
  480. Concolino D, Muzzi G, Pisaturo L, Piccirillo A, Di Natale P, Strisciuglio P. Precocious puberty in Sanfilippo IIIA disease: diagnosis and follow-up of two new cases. Eur J Med Genet. 2008;51(5):466-71.
  481. Ray LA, Eckert GJ, Eugster EA. Long-term experience with the use of a single histrelin implant beyond one year in patients with central precocious puberty. J Pediatr Endocrinol Metab. 2023;36(3):309-12.
  482. Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf). 2022;97(4):473-82.
  483. Akram M, Raza Rizvi SS, Qayyum M, Handelsman DJ. A classification of genes involved in normal and delayed male puberty. Asian J Androl. 2023;25(2):230-9.
  484. Vilain C, Mortier G, Van Vliet G, Dubourg C, Heinrichs C, de Silva D, et al. Hartsfield Holoprosencephaly-Ectrodactyly Syndrome in Five Male Patients: Further Delineation and Review. American Journal of Medical Genetics Part A. 2009;149a(7):1476-81.
  485. Pitteloud N, Meysing A, Quinton R, Acierno J, Dwyer A, Plummer L, et al. Mutations in fibroblast growth factor receptor 1 cause Kallmann syndrome with a wide spectrum of reproductive phenotypes. Molecular and Cellular Endocrinology. 2006:60-9.
  486. Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, et al. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology. 2009;150(6):2805-12.
  487. Williams KW, Sohn JW, Donato J, Lee CE, Zhao JJ, Elmquist JK, et al. The Acute Effects of Leptin Require PI3K Signaling in the Hypothalamic Ventral Premammillary Nucleus. J Neurosci. 2011;31(37):13147-56.
  488. Donato J, Cravo RM, Frazao R, Elias CF. Hypothalamic Sites of Leptin Action Linking Metabolism and Reproduction. Neuroendocrinology. 2011;93(1):9-18.
  489. Farooqi IS, O'Rahilly S. Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endoc. 2008;4(10):569-77.
  490. Tata B, Huijbregts L, Jacquier S, Csaba Z, Genin E, Meyer V, et al. Haploinsufficiency of Dmxl2, encoding a synaptic protein, causes infertility associated with a loss of GnRH neurons in mouse. PLoS Biol. 2014;12(9):e1001952.
  491. Margolin DH, Kousi M, Chan YM, Lim ET, Schmahmann JD, Hadjivassiliou M, et al. Ataxia, dementia, and hypogonadotropism caused by disordered ubiquitination. N Engl J Med. 2013;368(21):1992-2003.
  492. Synofzik M, Kernstock C, Haack TB, Schols L. Ataxia meets chorioretinal dystrophy and hypogonadism: Boucher-Neuhauser syndrome due to PNPLA6 mutations. J Neurol Neurosurg Psychiatry. 2015;86(5):580-1.
  493. Synofzik M, Gonzalez MA, Lourenco CM, Coutelier M, Haack TB, Rebelo A, et al. PNPLA6 mutations cause Boucher-Neuhauser and Gordon Holmes syndromes as part of a broad neurodegenerative spectrum. Brain. 2014;137(Pt 1):69-77.
  494. Pingault V, Bodereau V, Baral V, Marcos S, Watanabe Y, Chaoui A, et al. Loss-of-function mutations in SOX10 cause Kallmann syndrome with deafness. Am J Hum Genet. 2013;92(5):707-24.
  495. Tziaferi V, Kelberman D, Dattani MT. The role of SOX2 in hypogonadotropic hypogonadism. Sex Dev. 2008;2(4-5):194-9.
  496. Alatzoglou KS, Azriyanti A, Rogers N, Ryan F, Curry N, Noakes C, et al. SOX3 deletion in mouse and human is associated with persistence of the craniopharyngeal canal. J Clin Endocrinol Metab. 2014;99(12):E2702-8.
  497. Boehm U, Bouloux PM, Dattani MT, de Roux N, Dode C, Dunkel L, et al. Expert consensus document: European Consensus Statement on congenital hypogonadotropic hypogonadism--pathogenesis, diagnosis and treatment. Nat Rev Endocrinol. 2015;11(9):547-64.
  498. Joustra SD, Wehkalampi K, Oostdijk W, Biermasz NR, Howard S, Silander TL, et al. IGSF1 variants in boys with familial delayed puberty. Eur J Pediatr. 2015;174(5):687-92.
  499. Niederberger C. Re: Identification of HESX1 mutations in Kallmann syndrome. J Urol. 2014;191(4):1081.
  500. Kim HG, Kurth I, Lan F, Meliciani I, Wenzel W, Eom SH, et al. Mutations in CHD7, encoding a chromatin-remodeling protein, cause idiopathic hypogonadotropic hypogonadism and Kallmann syndrome. Am J Hum Genet. 2008;83(4):511-9.
  501. Marcos S, Sarfati J, Leroy C, Fouveaut C, Parent P, Metz C, et al. The Prevalence of CHD7 Missense Versus Truncating Mutations is Higher in Patients With Kallmann Syndrome than in Typical CHARGE Patients (vol 99, pg E2138, 2014). J Clin Endocr Metab. 2015;100(1):317-.
  502. Balasubramanian R, Choi JH, Francescatto L, Willer J, Horton ER, Asimacopoulos EP, et al. Functionally compromised CHD7 alleles in patients with isolated GnRH deficiency. P Natl Acad Sci USA. 2014;111(50):17953-8.
  503. Timmons M, Tsokos M, Asab MA, Seminara SB, Zirzow GC, Kaneski CR, et al. Peripheral and central hypomyelination with hypogonadotropic hypogonadism and hypodontia. Neurology. 2006;67(11):2066-9.
  504. Sato I, Onuma A, Goto N, Sakai F, Fujiwara I, Uematsu M, et al. A case with central and peripheral hypomyelination with hypogonadotropic hypogonadism and hypodontia (4H syndrome) plus cataract. J Neurol Sci. 2011;300(1-2):179-81.
  505. Pelletier F, Perrier S, Cayami FK, Mirchi A, Saikali S, Tran LT, et al. Endocrine and Growth Abnormalities in 4H Leukodystrophy Caused by Variants in POLR3A, POLR3B, and POLR1C. J Clin Endocrinol Metab. 2021;106(2):e660-e74.
  506. Liu S, Yan L, Zhou X, Chen C, Wang D, Yuan G. Delayed-onset adrenal hypoplasia congenita and hypogonadotropic hypogonadism caused by a novel mutation in DAX1. J Int Med Res. 2020;48(2):300060519882151.
  507. Muscatelli F, Strom TM, Walker AP, Zanaria E, Recan D, Meindl A, et al. Mutations in the Dax-1 Gene Give Rise to Both X-Linked Adrenal Hypoplasia Congenita and Hypogonadotropic Hypogonadism. Nature. 1994;372(6507):672-6.
  508. Jennings JE, Costigan C, Reardon W. Moebius sequence and hypogonadotrophic hypogonadism. Am J Med Genet A. 2003;123A(1):107-10.
  509. Alves C, Franco RR. Prader-Willi syndrome: endocrine manifestations and management. Arch Endocrinol Metab. 2020;64(3):223-34.
  510. Emerick JE, Vogt KS. Endocrine manifestations and management of Prader-Willi syndrome. Int J Pediatr Endocrinol. 2013;2013(1):14.
  511. Forsythe E, Beales PL. Bardet-Biedl syndrome. Eur J Hum Genet. 2013;21(1):8-13.
  512. Desai A, Jha O, Iyer V, Dada R, Kumar R, Tandon N. Reversible hypogonadism in Bardet-Biedl syndrome. Fertil Steril. 2009;92(1):391 e13-5.
  513. Jain V, Foo SH, Chooi S, Moss C, Goodwin R, Berland S, et al. Borjeson-Forssman-Lehmann syndrome: delineating the clinical and allelic spectrum in 14 new families. Eur J Hum Genet. 2023;31(12):1421-9.
  514. Wang T, Ren W, Fu F, Wang H, Li Y, Duan J. Digenic CHD7 and SMCHD1 inheritance Unveils phenotypic variability in a family mainly presenting with hypogonadotropic hypogonadism. Heliyon. 2024;10(1):e23272.
  515. Alavi O, Khamirani HJ, Zoghi S, Feili A, Dastgheib SA, Tabei SMB, et al. Two novel Warburg micro syndrome 1 cases caused by pathogenic variants in RAB3GAP1. Hum Genome Var. 2021;8(1):39.
  516. Kline AD, Moss JF, Selicorni A, Bisgaard AM, Deardorff MA, Gillett PM, et al. Diagnosis and management of Cornelia de Lange syndrome: first international consensus statement. Nat Rev Genet. 2018;19(10):649-66.
  517. Miraoui H, Dwyer AA, Sykiotis GP, Plummer L, Chung W, Feng BH, et al. Mutations in FGF17, IL17RD, DUSP6, SPRY4, and FLRT3 Are Identified in Individuals with Congenital Hypogonadotropic Hypogonadism. American Journal of Human Genetics. 2013;92(5):725-43.
  518. Bianco SDC, Kaiser UB. The genetic and molecular basis of idiopathic hypogonadotropic hypogonadism. Nature Reviews Endocrinology. 2009;5(10):569-76.
  519. Gonzalez-Martinez D, Kim SH, Hu YL, Guimond S, Schofield J, Winyard P, et al. Anosmin-1 modulates fibroblast growth factor receptor 1 signaling in human gonadotropin-releasing hormone olfactory neuroblasts through a heparan sulfate-dependent mechanism. J Neurosci. 2004;24(46):10384-92.
  520. Maione L, Albarel F, Bouchard P, Gallant M, Flanagan CA, Bobe R, et al. R31C GNRH1 mutation and congenital hypogonadotropic hypogonadism. Plos One. 2013;8(7):e69616.
  521. Bouligand J, Ghervan C, Tello JA, Brailly-Tabard S, Salenave S, Chanson P, et al. Isolated Familial Hypogonadotropic Hypogonadism and a GNRH1 Mutation. New Engl J Med. 2009;360(26):2742-8.
  522. Layman LC, Cohen DP, Jin M, Xie J, Li Z, Reindollar RH, et al. Mutations in gonadotropin-releasing hormone receptor gene cause hypogonadotropic hypogonadism. Nature Genetics. 1998;18(1):14-5.
  523. Topaloglu AK, Tello JA, Kotan LD, Ozbek MN, Yilmaz MB, Erdogan S, et al. Inactivating KISS1 mutation and hypogonadotropic hypogonadism. N Engl J Med. 2012;366(7):629-35.
  524. Xu C, Messina A, Somm E, Miraoui H, Kinnunen T, Acierno J, et al. KLB, encoding beta-Klotho, is mutated in patients with congenital hypogonadotropic hypogonadism. Embo Molecular Medicine. 2017;9(10):1379-97.
  525. Fergani C, Navarro VM. Expanding the Role of Tachykinins in the Neuroendocrine Control of Reproduction. Reproduction. 2016;153(1):R1-R14.
  526. Hanchate NK, Giacobini P, Lhuillier P, Parkash J, Espy C, Fouveaut C, et al. SEMA3A, a Gene Involved in Axonal Pathfinding, Is Mutated in Patients with Kallmann Syndrome. Plos Genetics. 2012;8(8).
  527. Abreu AP, Trarbach EB, de Castro M, Frade Costa EM, Versiani B, Matias Baptista MT, et al. Loss-of-function mutations in the genes encoding prokineticin-2 or prokineticin receptor-2 cause autosomal recessive Kallmann syndrome. J Clin Endocrinol Metab. 2008;93(10):4113-8.
  528. Dode C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P, Kottler ML, et al. Kallmann syndrome: mutations in the genes encoding prokineticin-2 and prokineticin receptor-2. PLoS Genet. 2006;2(10):e175.
  529. Kim HG, Ahn JW, Kurth I, Ullmann R, Kim HT, Kulharya A, et al. WDR11, a WD Protein that Interacts with Transcription Factor EMX1, Is Mutated in Idiopathic Hypogonadotropic Hypogonadism and Kallmann Syndrome. American Journal of Human Genetics. 2010;87(4):465-79.
  530. Saengkaew T, Ruiz-Babot G, David A, Mancini A, Mariniello K, Cabrera CP, et al. Whole exome sequencing identifies deleterious rare variants in CCDC141 in familial self-limited delayed puberty. NPJ Genom Med. 2021;6(1):107.
  531. Kotan LD, Hutchins BI, Ozkan Y, Demirel F, Stoner H, Cheng PJ, et al. Mutations in FEZF1 cause Kallmann syndrome. Am J Hum Genet. 2014;95(3):326-31.
  532. Lofrano-Porto A, Barra GB, Giacomini LA, Nascimento PP, Latronico AC, Casulari LA, et al. Luteinizing hormone beta mutation and hypogonadism in men and women. N Engl J Med. 2007;357(9):897-904.
  533. Layman LC, Porto AL, Xie J, da Motta LA, da Motta LD, Weiser W, et al. FSH beta gene mutations in a female with partial breast development and a male sibling with normal puberty and azoospermia. J Clin Endocrinol Metab. 2002;87(8):3702-7.
  534. Berger K, Souza H, Brito VN, d'Alva CB, Mendonca BB, Latronico AC. Clinical and hormonal features of selective follicle-stimulating hormone (FSH) deficiency due to FSH beta-subunit gene mutations in both sexes. Fertil Steril. 2005;83(2):466-70.
  535. Mancini A, Howard SR, Cabrera CP, Barnes MR, David A, Wehkalampi K, et al. EAP1 regulation of GnRH promoter activity is important for human pubertal timing. Human Molecular Genetics. 2019;28(8):1357-68.
  536. Styrkarsdottir U, Thorleifsson G, Sulem P, Gudbjartsson DF, Sigurdsson A, Jonasdottir A, et al. Nonsense mutation in the LGR4 gene is associated with several human diseases and other traits. Nature. 2013;497(7450):517-20.
  537. Zang S, Yin X, Li P. FTO-mediated m(6)A demethylation regulates GnRH expression in the hypothalamus via the PLCbeta3/Ca(2+)/CAMK signalling pathway. Commun Biol. 2023;6(1):1297.

Pediatric implications of normal insulin-GH-IGF-axis physiology

ABSTRACT

 

Understanding the involvement of the insulin-GH-IGF-axis in the different phases of human growth, development, and metabolism is the key to understanding human pathophysiology. The normal physiological actions of the axis optimize human growth and metabolism to impact adult height by approximately one third. IGF binding proteins modulate access of circulating IGF-I to the tissues and modulate IGF-I and -II access to the type 1 IGF receptor (IGF1R) at the cellular level. Complete lack of IGF1R signaling is most likely not compatible with a viable human fetus, while allelic haploinsufficiency impairs brain development and causes severe short stature. Lack of insulin receptor signaling in Leprechaunism may result in the rare event of an alive but severely small for gestational age baby that will only survive if treated with recombinant-IGF-1 to substitute inulin receptor signaling with IGF1R signaling via their common intracellular pathways. IGF-I gene defects result in mental retardation and severe fetal and postnatal growth failure with GH hypersecretion and marked insulin resistance. Likewise, IGF2 gene defects or imprinting defects cause severe fetal growth failure but somewhat less adverse effects on postnatal growth, more variable effects on brain development, and an absence of marked metabolic effects. GH fine-tunes insulin and IGF-I signaling with no impact on IGF-II expression and has a minor impact on fetal development and growth. GH effects on lipolysis are established in the newborn and ensure gluconeogenesis and prevents hypoglycemia after birth. The complete absence of GH expression such as in GHRHR or GH1 gene defects or absence of GH signaling in GHR or STAT5B gene defect leads to an adult height of 120-130cm if untreated, and has severe metabolic consequences. Even excess of insulin, GH, IGF-I and IGF-II signaling are associated with severe metabolic disease and excess growth and/or obesity. Malnutrition or malabsorption causes decreased insulin signaling which reduces GHR expression and blocks the GH signaling pathway leading to IGF-I expression (GHR uncoupling), while GH’s metabolic actions on lipolysis and gluconeogenesis are unaffected. GH signaling attenuate insulin actions on glucose metabolism which causes insulin resistance and hyperinsulinemia or may precipitate diabetes. However, insulin signaling pathways that enhances GHR function or suppress IGFBP-1 or SHBG production are still intact and promote anabolism, optimize growth, enhance androgen actions and play a mechanistic role in premature adrenarche and PCOS. Long-term nutritional deprivation compromises growth, while from a developmental perspective, decreased insulin signaling (leading to GHR uncoupling) prolongs life (at least in some experimental animal models) which ensures that fertile age is reached, and survival of the species is ensured. For the health of the general population, the subtle changes in insulin, GH and IGF-I signaling associated with gene polymorphisms or epigenetic changes programmed during fetal and early postnatal life and affecting gene expression are important. They determine growth and pubertal development in childhood and predispose the individual for developing the metabolic diseases and malignancies in adult life, as predicted by the Barker hypothesis. As the roles of the insulin-GH-IGF-axis in growth and metabolism, often discussed separately, are intimately linked they will be described jointly here.

 

EARLY WORK DEFINING THE INSULIN-GH-IGF-AXIS

 

Daughaday realized that the mitogenic effect of GH in the growth plate was not direct but mediated by Insulin-like Growth Factor-I (IGF-I), at that time named sulphation factor or somatomedin C (1). Another effect of IGF-I was insulin-like and not inhibited by insulin specific antibodies (2,3,4) and therefore it was named non-suppressible Insulin-like activity (NSILA). Hall and Van Wyk purified IGF-I from human muscle extracts (5,6) and realized that these biological activities originated from the same molecule. They also identified significant quantities in blood (7). The primary structure of IGF-I and Insulin-like Growth Factor-II (IGF-II) was discovered by Froesch and coworkers as a result of their persistent work to characterize the metabolic activity of NSILA (8,9). Soon after, the mitogenic activity of the sulphation factor or somatomedin C as well as somatomedin A was shown to be identical to IGF-I (10). Rechler and Nissley demonstrated that IGF-II was identical to multiplication stimulating activity, a factor known to stimulate DNA synthesis in chick embryo fibroblasts (11).

 

The concept that binding proteins existed for peptide hormones like the IGFs, similar to those for steroid and thyroid hormones, were suggested by studies from Zapf and Froesch (12) and by Hintz (13), demonstrating that NSILA was present in high molecular weight complexes in serum. The binding was exclusive to IGFs and did not apply to insulin or proinsulin despite their structural similarities. High molecular weight IGF-I complexes with IGFBPs were GH dependent (14) and formed a ternary complex composed of IGFBP-3 (15), the Acid Labile Subunit (ALS) (16) and IGF-I or IGF-II. Low molecular weight complexes contained IGF-I or IGF-II bound to an insulin regulated liver derived protein IGFBP-1 (17, 18), at first called the 28 kDa binding protein or PP12 (19). The existence of other IGF binding proteins, six in total, became clear when Hossenloop (20) developed Western ligand blotting as a technique to quantify these proteins. The components of the IGF-IGFBP-system are outlined in Figure 1.

 

Figure 1. The primary structures of IGF-I, IGF-II and insulin are similar. IGFs are produced by many differentiated cell types, and their bioactivity in the extracellular fluids or in the circulation are coordinated by six IGF binding proteins (IGFBP-1 through -6). IGFBP-3, the major binding protein in serum is stimulated by GH and it forms a large 150 kDa ternary complex with IGF-I or -II and the GH regulated acid labile subunit (ALS). IGFBP-5, an important supporter of bone tissue formation, also forms ternary complexes with IGF-I or -II and ALS. IGFBP-1, suppressed by insulin, is one of several binding proteins in the smaller 50 kDa binary complexes with IGF-I or –II. IGFBP-2 has inverse association with insulin under many physiological conditions. In contrast, IGFBP-4, -5 and -6 do not appear to be directly regulated by GH or insulin and are important local regulators of IGF activity in bone and the CNS. The type 1 IGF receptor (IGF1R) is the mediator of the mitogenic, anti-apoptotic, differentiating and metabolic effects of both IGF-I and -II. The structural similarity of the IGF1R with the insulin receptor (IR) explains the formation of hybrid receptors in cells that expresses both receptors such as myocytes and pre-adipocytes. Cross reactivity among the ligands and the receptors have been demonstrated, although it has minor importance under physiological conditions but may cause non-islet-cell tumor hypoglycemia due to unprocessed pro-IGFs with markedly decreased binding affinity to IGFBPs. A second receptor, exclusively binding IGF-II, work as a scavenger receptor and is identical to the mannose-6-phosphate receptor, known to direct proteins for degradation in the lysosomes. A second level of control of IGF bioactivity is exerted by IGFBP proteases which release IGF-I activity after fragmentation of IGFBPs. Specific production and regulation of IGFBP proteases at the tissue level controls processes such as ovulation and atherosclerosis. Furthermore, interaction of IGFBPs and IGFBP proteases with the extracellular matrix modify the binding affinity for the IGFs and are involved in prolonging the actions of IGFs at the tissue level. Extracellular matrix also signals though integrin receptors on the cell surface and modifies IGF-1R signaling. This figure also shows the existence of IGFBP-related proteins with markedly lower affinity for the IGFs and with physiological roles not related to their IGF binding.

 

ANIMAL EXPERIMENTS ESSENTIAL FOR THE UNDERSTANDING OF HUMAN INSULIN-GH-IGF AXIS PHYSIOLOGY

 

Insulin, IGF-I, and IGF-II and Their Receptors

 

Efstratiadis’ series of knock-outs of the insulin-GH-IGF-axis in mice in the early 1990s clearly confirmed its importance in fetal and postnatal growth and metabolism (21). It also predicted the phenotype of experiment of nature in humans with gene defects in the axis yet to be discovered. The studies opened new insights, not least the equal importance of IGF-I and IGF-II in fetal growth, reducing birth weight by about 60 % in both Igf1 knock-out (Igf1ko) and Igf2ko animals and demonstrating that the previous perceived concept that there was a fetal (IGF-II) and a postnatal (IGF-I) form of IGF was incorrect. IGF-I and -II had actions through the type 1 IGF receptor (IGF1R) demonstrated by Igf1rko animals with 45% of wild type birth weight and no further effect when crossed with Igf1ko animals. While Igf1ko animals were viable depending on genetic background and were non-fertile, the Igf1rko animals died from respiratory failure but with an absence of apparent malformations. Interestingly, crossing Igf2ko with Igf1rko resulted in further growth retardation indicating that IGF-II had actions through an additional receptor. Another new insight came from knock-out of the ‘mysterious type 2 IGF receptor’, identical to the mannose-6 phosphate receptor (M6P-R), specifically binding IGF-II and involved in internalization of proteins for lysosomal degradation. Knock-out of the Igf2r/M6pr resulted in increased serum and tissue levels of IGF-II and fetal overgrowth (140% of wild-type birth weight) (22). This receptor works to clear IGF-II and its presence in endothelial cells may, at least partly, explain the lack of endocrine actions of IGF-II due to its proteolytic lysosomal degradation (23). Thus, IGF-II effects on fetal growth are paracrine/autocrine actions mediated by the IGF1R. Knock-out of the Igf2r/M6pr gene combined with Igf2ko/Igf1rko could partly rescue growth retardation, a finding that was explained by IGF-II actions via the insulin receptor (INSR). The formation of heterodimers, more commonly named hybrid receptors, between type A or B isoforms of the insulin receptors (INSRA or INSRB) and the IGF1R of which IRA-IGF1R are highly expressed in the fetus (and in malignant cells) and activated by IGF-II, may further point to the importance of IGF-II during the fetal period. INSRB-IGF1R hybrids comprises up to 30 % of INSR and IGF-I receptors in muscle due to high expression of both and this hybrid predominantly responds to IGF-I (less to insulin) and explains the important role of IGF-I in growth and metabolism in skeletal muscle.

 

Postnatally, the Igf1ko mice continued to grow poorly, resulting in an adult weight 30% of wild-type and with poor gonadal function and delayed bone development. Knock-out of GH or its receptor (GHR), both expressed in the mouse fetus, did not affect birth size, indicating that the Igf1 gene is not under GH control during the fetal period. The actions of GH and its receptor on growth in mice were obvious from postnatal day 15 and largely slowed growth resulting in a 50 % reduction of wild-type adult weight. On the other hand, double Ghrko/Igf1ko resulted in further postnatal growth retardation relative to Igf1ko mice completely obstructing further weight gain after postnatal day 15 and supporting previous studies suggesting that progenitor cells in the growth plate require direct GH actions (24).

 

IGFBPs and IGFBP Proteases  

 

Like the above attempts to pinpoint the role of important ligands and receptors in the axis, steps to assess the role of IGF binding proteins involved in modulating IGF-I and IGF-II bioactivity were taken (reviewed by Pintar (25)). In contrast to the pronounced phenotypes caused by mutations in receptors and their ligands, the growth phenotypes of the various IGFBP knock-out animals were far less pronounced as were the metabolic changes observed (26,27,28). It was argued that there is a large degree of redundancy among the functions of the IGFBPs which to some extend contradicts their specialized functions in various tissues (29). However, this idea was to some extent supported by the finding of somewhat more pronounced phenotypes in double and triple knock-out animals (30). This is largely in accordance with the absence of reports of IGFBP gene defects causing growth retardation in humans. The most affected phenotype identified was that of Igfbp4ko mice who were growth retarded at birth and displayed poor postnatal growth (30). No such mutation has been identified in humans. IGFBP-4 is specifically degraded by the metalloproteinase PAPP-A (Pregnancy Associated Plasma Protease -A) produced by the placenta as well as bone and ovary. In Pappa knock-out animals a 20-30% reduction in body weight was reported (31). Interestingly, the growth restriction phenotype of mice null for Pappacould be rescued by disruption of IGF-II imprinting during embryonic development (32).

 

Endocrine Versus Paracrine Autocrine IGF-I

 

One of the controversies of the area has been the relative contribution to linear growth of circulating endocrine IGF-I largely produced by the liver versus peripherally produced IGF-I with major paracrine/autocrine actions on local tissues. The major importance of paracrine/autocrine IGF-I was demonstrated by liver specific Igf1ko mice (Ligf1ko) with largely unaffected longitudinal growth (33). Circulating levels were 20% of wild-type with compensatory elevation of GH, insulin resistance and hyperinsulinemia. With age the animals developed type 2 diabetes, underlining the metabolic consequences of largely elevated GH combined with circulating IGF-I deficiency (34). Somewhat unexpectedly, this animal model closely resembles children and adolescents with type 1 diabetes, as further elaborated on below.

 

Another model to assess the relative importance of endocrine versus paracrine/autocrine IGF-I is the liver-specific Ghrko mouse. It produces a similar phenotype but with more specific hepatic consequences of absent GH signaling (35).

 

INSULIN-GH-IGF AXIS PHYSIOLOGY, A PEDIATRIC PERSPECTIVE

 

Insulin-GH-IGF-Axis and Human Fetal Growth

 

IGF-I controls the pace of the cell cycle from early on in human embryogenesis. INSR and IGF1R is expressed in human pre-implantation blastocysts already from the 8-cell stage, while IGF-II is expressed already in the oocyte (36). After implantation, IGF-I is produced in the human embryo (37), but until then the source of IGF-I is thought to be the female reproductive tract, and it is known that the availability of the IGF-I ligand is important for blastocyst growth in human in vitro fertilization - IVF (38). IGF-I production is controlled by nutritional factors in the early embryo and even later during human fetal development (39). Circulating endocrine IGF-I increases with gestational age (40) and is strongly correlated with fetal growth in the second part of gestation (41,42). However, little has been reported concerning the regulatory control of the IGF1 gene in the human fetus. IGFBPs can be identified in the human fetal circulation (40), and recently the role of IGFBP-5 in regulating fetal growth was suggested by fetal growth retardation in the absence of a specific IGFBP-5 protease, PPAP-A2 (43). Insulin continues to be permissive for IGF-I production even after GH is established as the major pituitary stimuli controlling endocrine as well as paracrine/autocrine IGF-I, as described below.

 

Fetal Growth Restriction and Programming of the Insulin-GH-IGF-Axis Setpoint

 

Insulin resistance has been developmentally advantageous for mankind until very recent decades of excess food and sedentary life style. It was proposed by Barker et al (44) in his ‘fetal and infant origin of adult disease’ hypothesis that intrauterine restriction of growth compensated by excessive postnatal catch-up growth results in an increased risk of developing disease entities of the metabolic syndrome later in life. In his early epidemiological studies, he demonstrated that there is a U-shaped relationship between birth weight and risk of obesity, insulin resistance, type 1 diabetes, hypertension, dyslipidemia and ischemic heart disease, with lower birth weight (within the normal range) imposing a risk. Notably, at higher birth weights this risk rises again which may represent genetic risks of obesity and type 2 diabetes. The concept was that poor fetal nutrition would lower fetal IGF-I and program the child to a low IGF-I setpoint and slower postnatal growth, an epigenetic phenomenon that could be preserved over a few generations (45). At the same time, small for gestational age (SGA) babies becomes insulin resistant (46) and this trait is enforced by a low endocrine IGF-I setpoint (47), creating the best physiological circumstances for the storage of fat during short times of food availability in a world with limited access to food. However, in a world of plenty, this advantage would turn into a disadvantage and give fast increases in body weight, hyperinsulinemia, and the development of metabolic syndrome problems early on (reviewed by Dunger et al (48). New information even suggests that the parental nutritional state can impose epigenetic metabolic changes in the fetus (49).

 

Figure 2. In the fetus (insert) IGF-I increases with gestational age toward birth. Endocrine circulating IGF-I is strongly nutritionally dependent and correlated with birth size. Pituitary GH control of IGF-I production is not fully established during the first year of human life. The ability of serum IGF-I levels to increase during childhood is dependent on the shift from binary complexes of IGF-I with short half-life to a complete dominance of IGF-I bound in a stable ternary complex with the GH dependent proteins IGFBP-3 and ALS. Both these proteins increase, when pituitary GH control of the axis is established. During pubertal development, sex steroids change the set-point of negative IGF-I feedback and allow a peak of IGF-I in mid-puberty. Total IGF-I levels decline to low levels in senescence. Serum IGF-I reference values based on Juul (50).

 

Postnatal Establishment of Pituitary GH Control of IGF-I Production

 

In humans, full GH control of the IGFI gene, as well as the IGFBP3 and Acid Labile Subunit (ALS) genes, is developmentally regulated and established not until the first year of life. IGF-I and IGFBP-3 levels increase slowly from birth until a more rapid increase and peak during puberty, which is followed by a decline toward low levels in senescence (50, 51) (Figure 2). The late establishment of pituitary GH control of the axis is strongly supported by animal data from GHR KO mice reviewed above, and by a new model of Laron syndrome (GHR defect) in pigs (52). In accordance, newborns with mutations in the GH Releasing Hormone Receptor (GHRHR) gene resulting in an isolated GH deficiency (GHD) phenotype was associated with normal birth weight in one cohort (53) and slightly subnormal birth weight in another (54). In a subgroup of 12 children with congenital isolated GHD, birth weight (1·1 ± 0·8 SD) and length (0·5 ± 1·3 SD) was not affected (Mehta A). GH1 mutations appear to be slightly more affected with mean birth weights of −1.0 ± 0.9 (54). Studies of common polymorphisms in GH1 demonstrate dose effects of 150 and 100 grams in term newborns of normal and low birth weight, respectively (55).  Somewhat contradictory to the observations in animals, Savage et al (56) reported 27 prepubertal children with severe GH insensitivity syndrome (GHIS or Severe Primary IGF Deficiency (SPIGFD)) to have a median (range) birth weight SDS of -0.72 (1.75 - (-3.29)) and birth length SDS of -1.59 (0.63 - (-3.63)). SPIGFD in these patients were defined by phenotypic and biochemical characteristics and they were treated with recombinant human (rh)IGF-1 in one of the clinical trials leading to approval of this therapy, as described later. There was no complete genetic characterization of these patients and 7 patients had a normal serum GH Binding Protein (GHBP) suggesting that the extracellular part of GHR was not affected. In a monograph, Professor Zvi Laron (57), who gave his name to this syndrome, reported that birth weight is unaffected while birth length is slightly on the shorter side. In summary, human fetal growth is only marginally affected by GH. GH is detectable and the GHR is expressed in the human fetus and the metabolic effects of GH on lipolysis are essential to maintain normal levels of glucose in the newborn.

 

Given this critical role of GH in adjusting metabolism to the fasting condition, it is plausible that the metabolic effects of GH are required for optimal linear growth of the human fetus and that this explains marginal effects on linear growth of the human fetus. However, strict GH control of IGF-I in the human fetus would predict severe growth retardation of the above-mentioned genetic defects comparable with the birth size observed in defects in the IGF-I gene. And that is not observed: Children with IGF1 defects suffer from far more severe fetal growth retardation with birth weight SDS around -4 and birth length SDS of -5 to -6 (reviewed in (58). In the study by Mehta et al (Mehta A), children with congenital isolated GH deficiency demonstrated growth retardation as already at 6 months of age (−2·6 ± 1·0 SD and −2·2 ± 1·3 SD in weight and length, respectively) prior to starting treatment. This suggests that the developmental GH control of IGF-I production is established early after birth. It is in accordance with data from Jensen RB and Juul A et al (Jensen RB) suggesting that low IGF-I is a marker of GH deficiency early in life. Children with combined pituitary hormone deficiencies were even more growth retarded at 6 months (Metha A).

 

GHR Signaling Pathway to IGF1 Gene Transcription

 

The important cell signaling steps associated with GH stimulated IGF1 gene activation, transcription and IGF-I production are detailed in the Endotext chapter ‘Normal Physiology of Growth Hormone in Adults´. Briefly, GH binding to preformed dimeric GHR - JAK2 complexes introduces structural changes in the receptor complex that separates JAK2 inhibitory and kinase active sites and enables trans-phosphorylation of the two JAK2 molecules (reviewed in (59). The GHR belongs to the class 1 cytokine receptors which uses STAT as one of its principal secondary messengers, and the subsequent phosphorylation of two STAT5b molecules results in a phospo-STAT5b homodimer which translocate to the cell nucleus, binds to STAT5b recognition sites on the IGF1 gene promoter, and initiates transcription (Figure 3).

 

Primary and Secondary IGF-I Deficiency

 

IGF-I deficiency may be the result of low or inadequate production of GH. This condition is known as GH deficiency (GHD) but in analogy with other pituitary deficiencies leading to peripheral hormone deficiencies the term secondary IGF-I deficiency was proposed (Rosenfeld et al). GHD is severe in GH1 or GHRH-R gene defects and in some children with congenital GHD as well as after treatment of brain tumors with radiation therapy. However, these conditions are rare and most children treated with rhGH has less severe GHD or an indication not associated with GHD. Disorders of GH in childhood is outlined in the Endotext chapter Disorders of Growth Hormone in Childhood by Murray and Clayton (132)

 

Primary IGF-I deficiency or GH insensitivity is severe in homozygous genetic defects in genes including the GH receptor (Laron syndrome) gene, STAT-5b gene and IGF-I gene. Less severe growth retardation is reported in children with homozygous genetic defects in the ALS gene. Treatment with rhGH do not improve growth in these cases while rhIGF-1 is efficient in SPIGFD and approved by FDA and EMA. In many patients with severe primary IGF-I deficiency (SPIGFD) defined by low IGF-I (less than – 3 SDS or the 2.5th percentile), severe short stature (Height SDS less than – 3), normal GH secretion and absence of acquired insensitivity to GH (discussed below) genetic defects may be absent. Still treatment with rhIGF-1 may be as efficient as in patients with confirmed homozygous GHR defects (131).

 

Insulin Enhancement of GHR Signaling to IGF1 Gene Transcription

 

Insulin signaling enhances the GH signaling pathway to enable IGF-I production in the fed state and promotes linear growth and other anabolic responses (60, 61, 62). Moreover, GH signaling to elicit IGF1 gene transcription is blocked in the absence of appropriate insulin signaling, a phenomenon also known as un-coupling, and resulting in growth arrest (60, 61, 63, 64) (Figure 3). This is partly a result of insulin’s effects on hepatic GHR expression, and partly a post-receptor signaling effect as unraveled by extensive animal studies (reviewed in (60). In obese individuals with hyperinsulinemia, hepatic GHR expression is enhanced as indicated by elevated GHBP levels reflecting proteolytic cleavage of highly expressed surface GHR and release of the extracellular part to the circulation (65). This allows obese individuals to maintain normal serum IGF-I levels despite markedly diminished GH secretion (66, 67). Consequently, obese individuals have attenuated GH responses to GH secretagogues (68).

 

Figure 3. Multiple, partly identical, pathways have been described to be activated by the GHR, the INSR and many other hormone kinase receptors not shown on the slide. Limiting this cartoon to the GHR and INSR, still the complexity is very high and the potential candidate hubs for crosstalk are numerous. The crosstalk that, following activation of the GHR, leads to resistance to specific signaling events from the INSR (related to glucose metabolism) is more well established and describe in detail in the Endotext chapter ‘Normal Physiology of Growth Hormone in Adults´. In the current review the focus is on the crosstalk that is executed by activation of the INSR and results in enhanced signaling from the GHR leading to gene activation of IGF1 and other GH dependent genes such as IGFBP3 and ALS. There are basically no studies addressing this crosstalk on the cellular level despite the strong evidence for INSR signaling being permissive for IGF1 transcription. Given that mTORC1 and mTORC2, downstream the INSR, are essential hubs for substrate and energy sensing and thus controlling the switch between cell anabolism and catabolism, they appear to be strong candidates to determine whether IGF1 should be on or off. A further argument for their candidate role is the so far limited evidence of mTORC1 and mTORC2 involvement in branched chain amino acid sensing directly enhancing IGF1 transcription. The unique role of the Jak2, STAT5b pathway in connecting the GHR with IGF1 gene activation has not been challenged and is thus the major candidate pathway to be affected by INSR signaling crosstalk. It is less clear which of the signaling pathways from the GHR that results in enhanced lipolysis although the STAT5b pathways has been implicated. This is particularly interesting given that GHR induced lipolysis does not require INSR signaling crosstalk. The reader is encouraged to seek specific information regarding other GHR and INSR pathways depicted in the cartoon but not further discussed in this review.

 

While short-term fasting decreases serum IGF-I but does not affect GHBP (69), the triad of IGF-I deficiency, poor growth and pubertal delay/arrest in long term nutritional deficiency such as in anorexia nervosa is associated with low GHBP that is partly restored with weight gain (63). Also, circulating IGF-I deficiency due to hepatic under-insulinization in type 1 diabetes is associated with low GHBP levels. Normal circulating IGF-I and GHBP are fully restored only after experimental intra-peritoneal (70) or intraportal (71) insulin delivery.

 

Functional/Acquired IGF-I Deficiency - Uncoupling of GHR Signaling to IGF1 Gene Transcription and Maintained GHR Metabolic Signaling Due to Insulin Deficiency

 

Increased GHR signaling in obese children does not generally result in elevated IGF-I, due to negative feedback inhibition of GH. In contrast, impairment of GH signaling due to insulin deficiency cannot generally be compensated by GH hypersecretion. This is true in fasting children (63) and adults (62). The exact mechanisms by which insulin and GH signaling crosstalk on the post-receptor level is not yet understood (Figure 3). More recent data suggesting that FGF21 plays a role in mediating these events needs further confirmation (72). Interestingly, GH signaling leading to activation of lipolysis in adipose tissue and increased hepatic glucose production via both glycogenolysis and gluconeogenesis in the liver, is not affected by the absence of insulin crosstalk (reviewed in (72)). This has important implications in securing substrate mobilization and gluconeogenesis during fasting and explains the cardinal hypoglycemic symptoms in GHD and GHIS newborns in the absence of intrauterine growth retardation (IUGR). GH signaling leading to lipolysis is thought to involve STAT5b. Most information comes from animal models and involves GH signaling in the liver, but in mouse adipose tissue GHR KO downregulates beta-3 adrenergic receptor expression and inhibits lipolysis (73). GH effects are lost if STAT5b signaling is blocked (74), gene transcript profiles of GHR KO and STAT5b KO animals overlap largely, and STAT5b controls key regulator enzymes involved in lipid metabolism (75). However, if STAT5b mediates both metabolic signaling and IGF-I production it still needs to be understood where the two pathways diverge, and why GH metabolic signaling is not blocked in the absence of insulin crosstalk. In humans, recent studies have identified new GH signaling responses involving GH downregulation of fat-specific protein (FSP27), a negative regulator of lipolysis. MEK/ERK activation and inhibition of peroxisome proliferator-activated receptor-γ (PPARγ) are involved, and this offers an alternative signaling pathway from the GHR (76).

 

Interactions Among Endocrine Axes

 

The activity of the insulin-GH-IGF-axis is dependent on the other endocrine axes which have permissive actions on GH stimulated IGF-I expression and affect IGFBPs and proteases (Figure 4). For example, thyroxine is needed to enhance GH effects on endocrine IGF-I expression and a normal GH-IGF-IGFBP-axis is needed for optimal thyroid hormone production (77). Sex steroids further enhance the function of the GH-IGF-axis, most likely by attenuating pituitary and hypothalamic sensitivity to IGF-I negative feedback (78). The pivotal role of sex steroids on the setpoint of the axis is reflected by the peak circulating levels of IGF-I and IGFBP-3 reached in mid-puberty (50,51). On the other hand, GH via its stimulation of local IGF-I is important for testicular production of testosterone and spermatogenesis (79), and the local IGF-IGFBP-axis is involved in selection and growth of the primary follicle in the ovary, estradiol production and ovulation (80, 81). Finally, cortisol has impact on the actions of the GH-IGF-axis on growth by blocking IGF1R signaling leading to apoptosis (82) despite normal endocrine IGF-I levels (83).

 

Figure 4. Hypothalamic GH releasing hormone and somatostatin establish the pulsatile pituitary GH secretion that is established as the main regulator of endocrine and paracrine/autocrine IGF-I production during the first year of life in humans. Insulin is permissive for this regulation by modulating GHR signaling, and normal beta-cell release of insulin is required for normal liver derived endocrine IGF-I levels measured in serum (blue insert) that in most cases is a good marker of the local production and actions of IGF-I. During fasting the GH regulation of IGF-I is uncoupled, resulting in decreased IGF-I (and catabolism) and elevated GH secretion and maintained lipolytic signals securing gluconeogenesis and preventing hypoglycemia. Apart from insulin, the endocrine thyroid axis is important for normal GH induced IGF-I production and during pubertal development sex steroids from the gonads enhance the performance of the GH-IGF-axis presumable by relaxation of the negative IGF-I feedback on GH secretion allowing a higher set-point of the axis. Whether this is an estradiol effect is not fully elucidated but it is suggested by the fact that non-aromatizable androgens such as oxandrolone do not affect IGF-I levels. The actions of the adrenal axis are most likely local and involve actions on IGF1R signaling leading to apoptosis of stem cells in the growth plate and thus irreversible loss of height. Cortisol excess leaves endocrine IGF-I and GH levels largely unaffected.

 

Discordance Between Endocrine and Paracrine/Autocrine IGF-I

 

An important example of metabolic and mitogenic consequences of an unbalanced endocrine versus autocrine/paracrine insulin-GH-IGF-axis comes from observations in children and adolescents with type 1 diabetes (Figure 5). They suffer specifically from insulin deficiency in the hepatic portal circulation as a result of the subcutaneous delivery of insulin (reviewed by Dunger (64)). This attenuates their endocrine production of circulating IGF-I despite excessive GH secretion (84). Circulating IGF-I deficiency and GH hypersecretion induce insulin resistance which is further augmented by insufficient suppression of hepatic glucose output. To overcome this, higher subcutaneous insulin doses are needed to maintain glycemic control, and this results in aggravated systemic hyperinsulinemia. The importance of local tissue hyperinsulinemia and GH hypersecretion in generating high paracrine/autocrine IGF-I production and promoting mitogenic vascular events leading to diabetic long-term complications should not be underestimated. Based on this insight, a promising new drug targeting the alphaVbeta3 integrin affecting IGF-I signaling in smooth muscle cells has been found to inhibit the development of atherosclerotic lesions in diabetic pigs (85). Another consequence of a compensatory increased in local IGF-I activity is the finding of normal childhood and pubertal linear growth despite endocrine IGF-I deficiency in type 1 diabetes (86). It is interesting that the endocrine and paracrine/autocrine changes in the insulin-GH-IGF-axis observed in children with type 1 diabetes closely resembles those observed in liver IGF-I KO mice which eventually leads to diabetes in the KO mice. Given that portal delivery of insulin, which has the potential to completely restore IGF-I levels in type 1 diabetes (70, 71, 87), remains an experimental treatment, rhIGF-1 treatment to restore circulating IGF-I and suppress GH and decrease insulin needs appears to be the most feasible approach to take (88). In a 6-month clinical trial of a single daily injection of rhIGF-1 improved glycemic control in adolescents with type 1 diabetes were found (89). Long-term studies on diabetic vascular complications have yet to be performed.

 

If paracrine/autocrine IGF-I production is lost in addition to liver-derived IGF-I, the metabolic consequences become obvious. This situation was first reported in a boy with a deletion of exon 4 of the IGF-I gene (90) resulting in severe fetal and post-natal growth arrest, poor brain development and extreme insulin resistance with compensatory hyperinsulinemia and acanthosis nigricans. A short trial of treatment with rhIGF-1 resulted in normalization of circulating IGF-I, suppression of GH hypersecretion and a markedly decreased insulin response to a meal tolerance test (91). In this example and in type 1 diabetes, it has been discussed whether the normalization of glucose metabolism following rhIGF-I therapy is most importantly associated with insulin-like effects of IGF-I on glucose uptake in muscle or suppression of GH hypersecretion? Although most studies support the importance of GH suppression, prolonged actions of IGF-I similar to that of long-acting insulin analogs in type 1 diabetic patients are important. IGF-I is equipotent with insulin in stimulating glucose uptake in human muscle but has less effects in fat and liver (92). Reports that newborns with a complete lack of insulin effects due to inactivating defects in the INSR gene can now survive for extended time into adolescence when treated with recombinant IGF-I, that stimulate glucose uptake via the IGF1R sharing common signaling pathways with the INSR, support an important direct role of IGF-1 signaling on metabolism (93).

 

Figure 5. Changes in liver derived endocrine IGF-I measured in the circulation and paracrine/autocrine IGF-I are in most cases concordant. In the absence of practical and validated methods to measure IGF-I at the tissue site of action, paracrine/autocrine IGF-I activity is assessed by determining known physiological actions of IGF-I such as growth or glucose metabolism. Type 1 diabetes is a condition with discordant changes in endocrine vs. paracrine/autocrine changes in IGF-I that in many ways resembles those reported in a mouse model of conditional knock-out of IGF-1 expression in the liver. In type 1 diabetes, insulin deficiency in the liver, caused by a systemic rather than a portal insulin replacement therapy, results in a functional GHR signaling defect to IGF-I transcription (uncoupling). Low endocrine IGF-I production decreases circulating IGF-I and results in decreased negative pituitary feedback and GH hypersecretion. The lack of direct IGF-I effects on glucose uptake in muscle and the diabetogenic effects of GH (including maintained signaling to lipolysis) decreases insulin actions on glucose metabolism (known as insulin resistance). The portal insulin deficiency also fails to suppress hepatic glucose production. In other to maintain glycemic control, the increased insulin requirement can only be met by more subcutaneous insulin leading to systemic hyperinsulinemia. There is no direct information about local paracrine/autocrine IGF-I activity, but there are several indications that tissue hyperinsulinemia and GH hypersecretion results in a compensatory increase of tissue IGF-I activity. Firstly, linear growth is not impaired in children and adolescents with Type 1 Diabetes despite of their low endocrine IGF-I (comparable to levels in short stature children), indicating a compensatory upregulation of local IGF-I activity (IGF-I being the most important stimulator of longitudinal growth). Secondly, it is plausible that increased local IGF-I activity contributes to diabetes complications known to be tightly associated with increased rather than decreased IGF-I activity. While type 1 diabetes is not generally associated with increased risk of cancer, the increase in local production of IGF-I in obesity and type 2 diabetes may contribute.

 

Liver Disease and Endocrine Versus Paracrine/Autocrine IGF-1 Production

 

In children with severe liver disease, there may be similar discrepancies between circulating endocrine levels of IGF-I and IGF-I activity in peripheral levels contributed by paracrine/autocrine secretion of IGF-I (94). However, less is known about peripheral IGF-I activity and it is possible that there are more secondary metabolic and nutritional issues that could lower local IGF-I production and impact on linear growth. Particularly in liver cirrhosis associated with thalassemia major, which concomitantly can impair pituitary GH secretion, there is no secondary upregulation of local IGF-I and linear growth failure is common (95). Recently, increased IGF-I expression was reported in obese children with non-alcoholic fatty liver disease (NAFLD) and it was combined with upregulation of IGF1R (96), not expressed in the normal liver but involved in liver repair, such as after liver resection in a mouse model (97).

 

GH and Cytokine Crosstalk

 

STAT5b phosphorylation is also mediated by activation of other members of the cytokine receptor family and has an impact on immunological function: this is evident from the finding of immune deficient symptoms in children with STAT5b genetic defects (98) but these are not found in GHD and GHIS children.

 

Negative Control of GHR Signaling

 

Control of the GH signaling cascade is also under inhibitory control, principally by two mechanisms. Firstly, tyrosine phosphatases including PTP dephosphorylate GHR associated molecules. In Noonan’s syndrome genetic defects in the PTPN11 gene may affect this pathway and has been implicated in poor growth and poor response to GH therapy, although reports are conflicting (99, 100). In addition, the SOCS gene is activated by GH signaling and works as a short intracellular negative feedback loop which rapidly down-regulate GHR activity by internalization and receptor ubiquitinoylation resulting in lysosomal and proteasomal degradation (101).

 

Other Nutritional Signals to the GH-IGF-Axis

 

Nutritional supplementation increasing dietary protein intake from cow’s milk increases endocrine IGF-I, while an equal intake of animal protein from meat does not (102). It is possible that the amino-acid composition that differs depending on the dietary source may contribute, and there are other constituents in skimmed milk not present in meat. More likely, however, it is explained by a higher carbohydrate intake in the milk group (while fat content was higher with meat supplementation) and the finding that fasting insulin levels doubled (103), in accordance with the essential regulatory role of insulin on GHR signaling discussed above. There is, however, direct evidence for a regulatory role of amino-acids on IGF-I production that is independent of insulin. Branched chained amino-acids (BCAA) are known to stimulate cell growth by the activation of mTORC2, a protein complex that controls protein synthesis in cells by sensing nutrient and energy availability and is also one of the main signaling pathways of IGF-I and insulin (Figure 3). The role of BCAA has been studied in rats given a restricted diet containing high levels of BCAA, compared to a group given low levels of BCAA (104).

 

The availability of nutrients in the circulation might also have a direct effect on the production of IGF-I. Human breast adipocytes cultured in high glucose levels have been found to produce more IGF-I compared to adipocytes cultured in low glucose levels (105).

 

IGF-II

 

IGF-II is a paracrine/autocrine hormone which is as essential for fetal growth as IGF-I (21). IGF-II may also contribute considerable to postnatal growth although the absence of endocrine effects makes it difficult to study in humans. Less is known about the metabolic actions of IGF-II. Growth promoting actions of IGF-II is via the IGF1R and IGF1R-INSR hybrid receptors which has preference for IGF-II actions if the A isoform of the INSR receptor – expressed in the fetus and in malignantly transformed cells - pairs with the IGF1R in the hybrid (106). The human IGF2 gene is an imprinted gene (107) exclusively expressed from the paternal allele in certain tissues (reviewed by Rossignol (108). The imprinted promoter region is found in a complex configuration with the H19 gene on chromosome 11 and shares two important imprinting regions with this gene. Methylation of the imprinting region ICR1 results in expression of the paternal IGF2allele, while H19 gene expression is suppressed. Correct imprinting should lead to expression of the paternal allele only and sufficient expression of IGF-II for normal growth. Loss of methylation of the ICR1 on the paternal allele results in the phenotype of Silver Russell syndrome (SRS) which may also arise from other genetic aberrations that have not yet been linked to the IGF-II production or signaling cascade including maternal uniparental dyssomnia of chromosome 7.

 

SRS is characterized by proportional IUGR with severe SGA at birth, relative sparing of the brain with close to normal head circumference at birth, severe feeding difficulties in infancy and childhood (which in contrast to Prader-Willi syndrome does not rebound into feeding obsession later in life), postnatal growth retardation and body asymmetry. As indicated by the SRS phenotype, IGF2 gene transcription of some organs are not controlled by imprinting. The relative normal development of the brain and the relative macrocephaly of SRS is explained by the lack of imprinting control of IGF-II expression in the brain. Children with genetic mutations in the expressed paternal allele of the IGF2 gene, were reported to have an SRS phenotype. They had somewhat more pronounced psychomotor developmental problems compared with the SRS phenotype, which has increased risk of autism spectrum defects including attention deficit hyperactivity disorder. In SRS, there is a normal postnatal expression of the IGF2 gene in the liver leading to normal levels of circulating IGF-II. Interestingly, the normal endocrine levels of IGF-II do not overcome the postnatal growth restriction. A similar lack of endocrine IGF-II effects on growth and metabolism was reported in the IGF-I deficient child mentioned previously with a loss of exon 4 of the IGF1 gene. He had compensatory increased GH, IGFBP-3, ALS as a result of lack of negative IGF-I feedback and secondary to the increased IGF binding capacity, increased IGF-II (see also the chapter on IGF-binding proteins below). Another observation in favor of this view is the lack of a correlation between newborn cord levels of IGF-II and birth size (41). This contrasts with a strong positive correlation between cord blood IGF-I concentrations and birth size. The role of IGF-II should be viewed in the light of Efstradiadis series of knock-out experiments in mice (21) where the Igf2ko mice had the same degree of fetal growth retardation as the igf1ko which demonstrates that IGF-II is a paracrine/autocrine and not an endocrine hormone.

 

It is possible that circulating IGF-II after release from the ternary complex is cleared from the circulation by binding to the IGF2R – identical to the mannose-6-phosphate receptor – which is associated with lysosomes and results in degradation of IGF-II in endothelial cells (23). In the elegant mouse KO experiments by Efstradiadis et al (21), KO of the IGF2R resulted in fetal overgrowth. However, the largely elevated IGF-II serum levels in that model are more likely a secondary finding, while the lack of clearance of paracrine/autocrine IGF-II is the explanation for the excessive growth.

 

IGF Binding Proteins

 

Six IGFBPs bind IGF-I and IGF-II inside and outside the circulation and has impact on IGF bioactivity (reviewed by Clemmons (109). The IGFBP-related proteins share some structural similarities with the six IGFBPs but have no relevant impact on IGF bioactivity. GF-I passes the endothelium intact primarily via IGF1R mediated transcytosis and this process is essential for endocrine actions of liver derived IGF-I (110). Limited experimental evidence from animal and tissue cultures suggest that IGF-I complexed with IGFBP-1 and -2 may leave the circulation, although the extent and importance is unclear (Bar 1990).  After endothelial passage IGF-I redistributes to soluble IGFBPs in the extravascular fluids or IGFBPs bound on extracellular matrix or cell surfaces (111). The concentration of unbound IGF-I in the circulation is likely to be proportional to unbound IGF-I concentrations in the tissues, but they are not equal and may have different relationship in different target tissues with differentially-expressed IGFBPs.

 

As pointed out earlier in this review, transgenic animal studies disrupting one or more IGFBPs have not suggested that a marked growth phenotype should be expected in children and no IGFBP mutations causing growth retardation in children had been reported to the best of my knowledge. Interestingly, as predicted by the Igfbp4-ko and the pappa-ko animal models described previously, a human PAPP-A2 gene defect with growth phenotype was recently reported as detailed below (43).

 

Free IGF-I Assays

 

Assays claiming to measure free circulating IGF-I have been developed, but it is unclear to what extent different techniques are influenced by redistribution of IGF-I among IGFBPs associated with the assay procedure (112). Anyway, the fact that IGF-I redistribute among extravascular IGFBPs after passing the endothelium is likely to affect the local tissue bioavailability even more. Moreover, the fact that most data in the literature originate from one assay technique established in one single laboratory has resulted in a lack of confirmatory reports. In a few cases, measurements with different free IGF-I assays have been reported from the same study/experiment with large differences in results (113). The bottom line is that measurements of free IGF-I have not been demonstrated to better predict different physiological or pathophysiological conditions in humans and do therefore not provide any clinically important contributions (114, 112). Techniques to assess IGF-I at the tissue site of action pose practical and methodological challenges. Attempts to establish and validate a method to determine local tissue levels by microdialysis have been reported in adolescents with type 1 diabetes, where endocrine levels are a poor marker of local IGF-I activity (115).

 

Ternary Complex Formation

 

The developmental establishment of GH control over the IGF1, IGFBP3 and IGFALS genes in early childhood initiates the dominance of the ternary complex formed by IGF-I or IGF-II and IGFBP-3 (or IGFBP-5) and ALS as the quantitatively most important circulating form of IGF-I and IGF-II (reviewed by Baxter (116). In the human fetus and newborn, serum IGFBP-3 and ALS concentrations are low and ternary complex formation is absent (117). Although IGF2 gene expression is not under GH control, the circulating levels of IGF-II are largely influenced by GH status since IGF-II (as well as IGF-I and IGFBP-3) is rapidly cleared from the circulation if not bound in the ternary complex. This can be observed in children with SPIGFD, who are deficient in IGF-I as well as IGFBP-3 and ALS, and in whom sc injected rhIGF-I displays a very fast serum clearance rate (118). As mentioned, formation of the ternary complex also governs the circulating levels of IGFBP-3 which under physiological conditions is present in a 1:1 molar relationship with IGF-I plus IGF-II. ALS is a large glucoprotein that under physiological conditions are present in a two-fold molar excess (16).

 

Immunometric IGFBP-3 assays have been claimed to be more predictive of GH status in very young children; however, the support for that is weak. It is rather a misconception related to problems of commercial IGF-I assays at the lower end of IGF-I detection. Moreover, IGFBP-3 has been claimed to provide information about IGF-I bioavailability from calculating the molar ratio of total IGF-I to IGFBP-3. Given that both IGF-I and IGFBP-3 are rapidly cleared from the circulation if unbound, using the IGF-I/IGFBP-3 ratio and disregarding IGF-II concentrations (that are 2-3-fold those of IGF-I on a molar basis) does not make any sense. During puberty, for example, IGF-I bioactivity is increased (114). This is dependent on the 3-4-fold increase in total IGF-I (50), which consequently results in an increase in unbound IGF-I, even if the increase is matched with the same absolute molar increase in IGFBP-3 (and complexed with ALS in a ternary complex). A common view is that increased IGF-I bioactivity depends on a higher IGF-I/IGFBP-3 molar ratio during puberty. However, the increase in molar ratio is entirely explained by the fact that IGF-I and IGFBP-3 increase with the same number of moles per liter, but with a larger relative increase in IGF-I than IGFBP-3 and with IGF-II molar concentrations being unchanged (112).

 

IGFBP Proteolysis and Physiological Consequences

 

The fact that proteolytic cleavage of IGFBP-3 is common, and may result in falsely elevated IGFBP-3 immunoactivity, is the most likely reason for observing a low IGF-I/ IGFBP-3 ratio. Under certain physiological conditions first described in pregnancy (119, 120), specific proteases cleave IGFBP-3 into several proteolytic fragments of which each may retain immunoactivity and thus give rise to signals in an immunometric assay (121). This will lead to overestimations of the IGFBP-3 immunoreactivity in pregnancy, which is already truly increased due to increased placental GH tonus. It may also lead to the erroneous conclusion that IGF-I bioactivity is decreased. On the contrary, IGF-I bioactivity is increased in the maternal circulation resulting from increased total serum IGF-I and decreased binding affinity of fragmented IGFBP-3 (122). There is strong experimental evidence that IGFBP proteolysis results in lower IGF binding affinity. The finding that partial IGFBP-3 proteolysis, such as in pregnancy, does not disrupt the ternary complex, has questioned its significance. However, evidence for increased IGF-I bioactivity in a ternary complex with fragmented IGFBP-3 exists (123). IGFBP-3 proteolysis has also been described in insulin resistant states such as fasting, obesity and type 1 and 2 diabetes (62, 124, 125). While several known proteolytic enzymes such as those involved in blood clotting (126) and cancer metastasis (127, 128) have been identified as IGFBP-3 proteases, the identity of the pregnancy protease is still not resolved.

 

Recently, a human gene mutation of PAPP-A2, a circulating and tissue protease with IGFBP-5 (and to some extent IGFBP-3) as its primary substrate (Gaudamauskes et al), was demonstrated to have a marked growth phenotype involving fetal and post-natal growth retardation in children in a consanguineous family (43). Largely elevated levels of circulating IGF-I but as a result of absence of proteolysis of IGFBP-5 and IGFBP-3, necessary for disruption of ternary complex formation, IGF-I bioactivity in serum is low and presumably tissue bioactivity of IGF-I (and IGF-II) is low. Functionally, this is a state of severe primary IGF-I deficiency (despite of elevated total serum IGF-I) and pharmacokinetic studies suggested that sc. Injections of rhIGF-1 resulted in a fraction of unbound IGF-I in serum despite the impaired proteolysis of IGFBP-5 and IGFBP-3 (133). Attempts to improve linear growth by rhIGF-I treatment has been reported to result in some improvements in a few affected children but not all (134, 135)

 

It is beyond the scope of this chapter to review the overwhelming evidence from cell biology experiments demonstrating the important role of IGFBPs in modulating IGF bioactivity and the role of IGFBP proteases and their actions at the cellular level. Furthermore, IGFBPs other than IGFBP-3 may play a role in the access of IGF-I to various tissues (129).

 

SUMMARY

 

In the present review the pivotal role of nutrition and insulin in determining the regulation and actions of the GH-IGF-axis is reviewed. For the pediatrician, caring for patients in a phase of rapid growth and development, it is important to refer to normality and understand the requirements for a normal insulin-GH-IGF-axis in order to succeed in this task. In the complex work-up, treatment and management of growth disorders a thorough understanding of the normal physiology of the axis is essential in taking the right actions (130, 131). From the normal physiology of this axis, it is possible to understand the consequences of various genetic defects and disorders that affect its regulation and function. The most severe conditions associated with defects in the axis may cause a loss of adult height of approximately 1/3 and may cause severe developmental and neurological deficits and compromise pubertal maturation and fertility. Minor changes in the setpoint of the axis caused by programming of the fetus exposed to intra-uterine growth retardation may predispose the individual for poor linear growth and later metabolic disease, insights that the pediatrician should be aware of and consider in order to improve health and prevent later disease.

 

REFERENCES

 

  1. Salmon WD, Daughaday WH. A hormonally controlled serum factor which stimulates sulfate incorporation by cartilage in vitro. J Lab Clin Med. 1957;49:825–826.
  2. Leonards JR. Insulin-like activity of blood, what is it? Federation Proc. 1959;18:272.
  3. Froesch ER, Bürgi WA, Ramseier EB, Bally P, Labhart A. Antibody-suppressible and nonsuppressible insulin-like activities in human serum and their physiological significance. An insulin assay with adipose tissue of increased precision and specificity. J Clin Invest 1963;42:1816– 1834.
  4. Froesch ER, Muller WA, Bürgi H, Waldvogel M, Labhart A. Non-suppressible insulin-like activity of human serum. II. Biological properties of plasma extracts with non-suppressible insulin-like activity. Biochim Biophys Acta. 1966;121:360–374.
  5. Van Wyk JJ, Hall K, Van den Brande JL, Weaver RP. Further purification and characterization of sulfation factor and thymidine factor from acromegalic plasma. J Clin Endocrinol Metab. 1971;32:389–403.
  6. Van Wyk JJ, Hall K, Weaver RP. Partial purification of sulphation factor and thymidine factor from plasma. Biochim Biophys Acta 1969;192:560–562.
  7. Uthne K. Human Somatomedin. Purification and some studies on their biological actions. Thesis, Stockholm, 1973.
  8. Rinderknecht E, Humbel RE. The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin. J Biol Chem. 1978;253:2769–2776.
  9. Rinderknecht E, Humbel RE. Primary structure of human insulin-like growth factor II. Febs Letters. 1978;89:283–286.
  10. Klapper DG, Svoboda ME, Van Wyk JJ. Sequence analysis of somatomedin-C: confirmation of identity with insulin-like growth factor I. Endocrinology. 1983;112:2215–2217.
  11. Moses AC, Nissley SP, Short PA, Rechler MM, Podskalny JM. Purification and characterization of multiplication stimulating activity. Insulin- like growth factors purified from rat-liver- cell-conditioned medium. Eur J Biochem. 1980;103:387–400.
  12. Zapf J, Waldvogel M, Froesch ER. Binding of nonsuppressible insulinlike activity to human serum: evidence for a carrier protein. Arch Biochem Biophys. 1975;169:638–645.
  13. Hintz RL, Liu F. Demonstration of specific plasma protein binding sites for somatomedin. J Clin Endocrinol Metab. 1977;45:988– 495.
  14. Hintz RL, Liu F. Demonstration of specific plasma protein binding sites for somatomedin. J Clin Endocrinol Metab. 1977;45:988– 495.
  15. Martin JL, Baxter RC. Insulin-like growth factor-binding protein from human plasma. Purification and characterization. J Biol Chem. 1986;261:8754–8760.
  16. Baxter RC. Characterization of the acid-labile subunit of the growth hormone-dependent insulin-like growth factor binding protein complex. J Clin Endocrinol Metab. 1988;67:265–272.
  17. Póvoa G, Enberg G, Jörnvall H, Hall K. Isolation and characterization of a somatomedin-binding protein from mid-term human amniotic fluid. Eur J Biochem. 1984;144:199-204.
  18. Póvoa G, Roovete A, Hall K. Cross-reaction of serum somatomedin-binding protein in a radioimmunoassay developed for somatomedin-binding protein isolated from human amniotic fluid. Acta Endocrinol (Copenh). 1984;107:563-70.
  19. Suikkari AM, Koivisto VA, Rutanen EM, Yki Jarvinen H, Karonen SL, Seppala M. Insulin regulates the serum levels of low molecular weight insulin-like growth factor-binding protein. J Clin Endocrinol Metab. 1988;66:266–272.
  20. Hossenlopp P, Seurin D, Segovia B, Quinson B, Hardouin S, Binoux M. Analysis of serum insulin-like growth factor binding proteins using Western competitive binding studies. Anal Biochem 1986;154:138–143.
  21. Efstratiadis A. Genetics of mouse growth. Int J Dev Biol. 1998;42:955-976.
  22. Ludwig T, Eggenschwiler J, Fisher P, D'Ercole AJ, Davenport ML, Efstratiadis A. Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null backgrounds. Dev Biol. 1996;177:517-535.
  23. Hachiya HL, Carpentier JL, King GL. Comparative studies on insulin-like growth factor II and insulin processing by vascular endothelial cells. Diabetes. 1986;35:1065-1072.
  24. Lindahl A, Isgaard J, Carlsson L, Isaksson OG. Differential effects of growth hormone and insulin-like growth factor I on colony formation of epiphyseal chondrocytes in suspension culture in rats of different ages. Endocrinology. 1987;121:1061-1069.
  25. Pintar JE, Schuller A, Cerro JA, Czick M, Grewal A, Green B. Genetic ablation of IGFBP-2 suggests functional redundancy in the IGFBP family. Prog Growth Factor Res. 1995;6:437-45.
  26. Wood TL, Rogler LE, Czick ME, Schuller AG, Pintar JE. Selective alterations in organ sizes in mice with a targeted disruption of the insulin-like growth factor binding protein-2 gene. Mol Endocrinol. 2000;14:1472-82.
  27. Murali SG, Liu X, Nelson DW, Hull AK, Grahn M, Clayton MK, Pintar JE, Ney DM. Intestinotrophic effects of exogenous IGF-I are not diminished in IGF binding protein-5 knockout mice. Am J Physiol Regul Integr Comp Physiol. 2007;292:R2144-2150.
  28. Ning Y, Hoang B, Schuller AG, Cominski TP, Hsu MS, Wood TL, Pintar JE. Delayed mammary gland involution in mice with mutation of the insulin-like growth factor binding protein 5 gene. Endocrinology. 2007;148:2138-2147.
  29. Pintar JE, Cerro JA, Wood TL. Genetic approaches to the function of insulin-like growth factor-binding proteins during rodent development. Horm Res. 1996;45:172-177.
  30. Ning Y, Schuller AG, Bradshaw S, Rotwein P, Ludwig T, Frystyk J, Pintar JE. Diminished growth and enhanced glucose metabolism in triple knockout mice containing mutations of insulin-like growth factor binding protein-3, -4, and -5. Mol Endocrinol. 2006;20:2173-2186.
  31. Conover CA, Bale LK, Overgaard MT, Johnstone EW, Laursen UH, Füchtbauer EM, Oxvig C, van Deursen J. Metalloproteinase pregnancy-associated plasma protein A is a critical growth regulatory factor during fetal development. Development. 2004;131:1187-1194.
  32. Bale LK, Conover CA. Disruption of insulin-like growth factor-II imprinting during embryonic development rescues the dwarf phenotype of mice null for pregnancy-associated plasma protein-A. J Endocrinol. 2005;186:325-331.
  33. Yakar S, Liu JL, Stannard B, Butler A, Accili D, Sauer B, LeRoith D. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci U S A. 1999;96:7324-7329.
  34. Yu R, Yakar S, Liu YL, Lu Y, LeRoith D, Miao D, Liu JL. Liver-specific IGF-I gene deficient mice exhibit accelerated diabetes in response to streptozotocin, associated with early onset of insulin resistance. Mol Cell Endocrinol. 2003;204:31-42.
  35. Fan Y, Menon RK, Cohen P, Hwang D, Clemens T, DiGirolamo DJ, Kopchick JJ, Le Roith D, Trucco M, Sperling MA. Liver-specific deletion of the growth hormone receptor reveals essential role of growth hormone signaling in hepatic lipid metabolism. J Biol Chem. 2009;284:19937-19944.
  36. Lighten AD, Hardy K, Winston RM, Moore GE. Expression of mRNA for the insulin-like growth factors and their receptors in human preimplantation embryos. Mol Reprod Dev. 1997;47:134-9.
  37. Han VK, D'Ercole AJ, Lund PK. Cellular localization of somatomedin (insulin-like growth factor) messenger RNA in the human fetus. Science. 1987;236:193-197.
  38. Yu Y, Yan J, Li M, Yan L, Zhao Y, Lian Y, Li R, Liu P, Qiao J. Effects of combined epidermal growth factor, brain-derived neurotrophic factor and insulin-like growth factor-1 on human oocyte maturation and early fertilized and cloned embryo development. Hum Reprod. 2012;27:2146-59.
  39. Rifas-Shiman SL, Fleisch A, Hivert MF, Mantzoros C, Gillman MW, Oken E, Lighten AD. First and second trimester gestational weight gains are most strongly associated with cord blood levels of hormones at delivery important for glycemic control and somatic growth. Metabolism. 2017;69:112-119.
  40. Bang P, Westgren M, Schwander J, Blum WF, Rosenfeld RG, Stangenberg M. Ontogeny of insulin-like growth factor-binding protein-1, -2, and -3: quantitative measurements by radioimmunoassay in human fetal serum. Pediatr Res. 1994;36:528-536.
  41. Lassarre C, Hardouin S, Daffos F, Forestier F, Frankenne F, Binoux M. Serum insulin-like growth factors and insulin-like growth factor binding proteins in the human fetus. Relationships with growth in normal subjects and in subjects with intrauterine growth retardation. Pediatr Res. 1991;29:219-225.
  42. Östlund E, Bang P, Hagenäs L, Fried G. Insulin-like growth factor I in fetal serum obtained by cordocentesis is correlated with intrauterine growth retardation. Hum Reprod. 1997;12:840-844.
  43. Dauber A, Muñoz-Calvo MT, Barrios V, Domené HM, Kloverpris S, Serra-Juhé C, Desikan V, Pozo J, Muzumdar R, Martos-Moreno GÁ, Hawkins F, Jasper HG, Conover CA, Frystyk J, Yakar S, Hwa V, Chowen JA, Oxvig C, Rosenfeld RG, Pérez-Jurado LA, Argente J. Mutations in pregnancy-associated plasma protein A2 cause short stature due to low IGF-I availability. EMBO Mol Med. 2016;8:363-374.
  44. Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1:1077-1081.
  45. Tobi EW, Goeman JJ, Monajemi R, Gu H, Putter H, Zhang Y, Slieker RC, Stok AP, Thijssen PE, Müller F, van Zwet EW, Bock C, Meissner A, Lumey LH, Eline Slagboom P, Heijmans BT. DNA methylation signatures link prenatal famine exposure to growth and metabolism. Nat Commun. 2014;5:5592.
  46. Veening MA, Van Weissenbruch MM, Delemarre-Van De Waal HA. Glucose tolerance, insulin sensitivity, and insulin secretion in children born small for gestational age. J Clin Endocrinol Metab. 2002;87:4657-4661.
  47. Sandhu MS, Heald AH, Gibson JM, Cruickshank JK, Dunger DB, Wareham NJ. Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet. 2002;359:1740-1745.
  48. Dunger D, Yuen K, Ong K. Insulin-like growth factor I and impaired glucose tolerance. Horm Res. 2004;62 Suppl 1:101-107.
  49. Öst A, Lempradl A, Casas E, Weigert M, Tiko T, Deniz M, Pantano L, Boenisch U, Itskov PM, Stoeckius M, Ruf M, Rajewsky N, Reuter G, Iovino N, Ribeiro C, Alenius M, Heyne S, Vavouri T, Pospisilik JA. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell. 2014;159:1352-1364.
  50. Juul A, Bang P, Hertel NT, Main K, Dalgaard P, Jørgensen K, Müller J, Hall K, Skakkebaek NE. Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, sex, stage of puberty, testicular size, and body mass index. J Clin Endocrinol Metab. 1994;78:744-752.
  51. Juul A, Dalgaard P, Blum WF, Bang P, Hall K, Michaelsen KF, Müller J, Skakkebaek NE. Serum levels of insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3) in healthy infants, children, and adolescents: the relation to IGF-I, IGF-II, IGFBP-1, IGFBP-2, age, sex, body mass index, and pubertal maturation. J Clin Endocrinol Metab. 1995;80:2534-42.
  52. Hinrichs A, Kessler B, Kurome M, Blutke A, Kemter E, Bernau M, Scholz AM, Rathkolb B, Renner S, Bultmann S, Leonhardt H, de Angelis MH, Nagashima H, Hoeflich A, Blum WF, Bidlingmaier M, Wanke R, Dahlhoff M, Wolf E. Growth hormone receptor-deficient pigs resemble the pathophysiology of human Laron syndrome and reveal altered activation of signaling cascades in the liver. Mol Metab. 2018;11:113-128
  53. Alba M, Salvatori R. Familial Growth Hormone Deficiency and Mutations in the GHRH Receptor Gene. Vitam Horm. 2004;69:209-220.
  54. Alatzoglou KS, Turton JP, Kelberman D, Clayton PE, Mehta A, Buchanan C, Aylwin S, Crowne EC, Christesen HT, Hertel NT, Trainer PJ, Savage MO, Raza J, Banerjee K, Sinha SK, Ten S, Mushtaq T, Brauner R, Cheetham TD, Hindmarsh PC, Mullis PE, Dattani MT. Expanding the spectrum of mutations in GH1 and GHRHR: genetic screening in a large cohort of patients with congenital isolated growth hormone deficiency. J Clin Endocrinol Metab. 2009;94:3191-3199.
  55. The role of growth hormone in determining birth size and early postnatal growth, using congenital growth hormone deficiency (GHD) as a model. Mehta A, Hindmarsh PC, Stanhope RG, Turton JP, Cole TJ, Preece MA, Dattani MT.Clin Endocrinol (Oxf). 2005 Aug;63(2):223-31. doi: 10.1111/j.1365-2265.2005.02330.x.
  56. Savage MO, Blum WF, Ranke MB, Postel-Vinay MC, Cotterill AM, Hall K, Chatelain PG, Preece MA, Rosenfeld RG. Clinical features and endocrine status in patients with growth hormone insensitivity (Laron syndrome). J Clin Endocrinol Metab. 1993;77:1465-1471.
  57. Laron Z. Lessons from 50 years of study of Laron Syndrome. Endocr Pract. 2015;21:1395-1402. Jensen RB, Jeppesen KA, Vielwerth S,  Michaelsen KM,  Main KM,  Skakkebaek NE, Juul Insulin-like growth factor I (IGF-I) and IGF-binding protein 3 as diagnostic markers of growth hormone deficiency in infancy Horm Res 2005;63(1):15-21. doi: 10.1159/000082456.Epub 2004 Nov 30.
  58. Waters MJ, Hoang HN, Fairlie DP, Pelekanos RA, Brown RJ. New insights into growth hormone action. J Mol Endocrinol. 2006;36:1-7.
  59. Maes M, Maiter D, Thissen JP, Underwood LE, Ketelslegers JM. Contributions of growth hormone receptor and postreceptor defects to growth hormone resistance in malnutrition. Trends Endocrinol Metab. 1991;2:92-97.
  60. Argente J, Caballo N, Barrios V, Pozo J, Muñoz MT, Chowen JA, Hernández M. Multiple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in prepubertal children with exogenous obesity: effect of short- and long-term weight reduction. J Clin Endocrinol Metab. 1997;82:2076-2083.
  61. Bang P, Brismar K, Rosenfeld RG, Hall K. Fasting affects serum insulin-like growth factors (IGFs) and IGF-binding proteins differently in patients with noninsulin-dependent diabetes mellitus versus healthy nonobese and obese subjects. J Clin Endocrinol Metab. 1994;78:960-967.
  62. Argente J, Caballo N, Barrios V, Muñoz MT, Pozo J, Chowen JA, Morandé G, Hernández M. Multiple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in patients with anorexia nervosa: effect of short- and long-term weight recuperation. J Clin Endocrinol Metab. 1997;82:2084-2092.
  63. Dunger DB, Acerini CL. IGF-I and diabetes in adolescence. Diabetes Metab. 1998;24:101-107.
  64. Frystyk J, Skjaerbaek C, Vestbo E, Fisker S, Orskov H. Circulating levels of free insulin-like growth factors in obese subjects: the impact of type 2 diabetes. Diabetes Metab Res Rev. 1999;15:314-322.
  65. Van Dam EW, Roelfsema F, Helmerhorst FH, Frölich M, Meinders AE, Veldhuis JD, Pijl H. Low amplitude and disorderly spontaneous growth hormone release in obese women with or without polycystic ovary syndrome. J Clin Endocrinol Metab. 2002;87:4225-4230.
  66. Roemmich JN, Clark PA, Weltman A, Veldhuis JD, Rogol AD. Pubertal alterations in growth and body composition: IX. Altered spontaneous secretion and metabolic clearance of growth hormone in overweight youth. Metabolism. 2005;54:1374-1383.
  67. Stanley TL, Levitsky LL, Grinspoon SK, Misra M. Effect of body mass index on peak growth hormone response to provocative testing in children with short stature. J Clin Endocrinol Metab. 2009;94:4875-4881.
  68. Salgin B, Marcovecchio ML, Hill N, Dunger DB, Frystyk J. The effect of prolonged fasting on levels of growth hormone-binding protein and free growth hormone. Growth Horm IGF Res. 2012;22:76-81.
  69. Hanaire-Broutin H, Sallerin-Caute B, Poncet MF, Tauber M, Bastide R, Chalé JJ, Rosenfeld R, Tauber JP. Effect of intraperitoneal insulin delivery on growth hormone binding protein, insulin-like growth factor (IGF)-I, and IGF-binding protein-3 in IDDM. Diabetologia. 1996;39:1498-1504.
  70. Shishko PI, Dreval AV, Abugova IA, Zajarny IU, Goncharov VC. Insulin-like growth factors and binding proteins in patients with recent-onset type 1 (insulin-dependent) diabetes mellitus: influence of diabetes control and intraportal insulin infusion. Diabetes Res Clin Pract. 1994;25:1-12.
  71. Chia DJ. Mechanisms of growth hormone-mediated gene regulation. Mol Endocrinol. 2014;28:1012-1025.
  72. Heffernan M1, Summers RJ, Thorburn A, Ogru E, Gianello R, Jiang WJ, Ng FM. The effects of human GH and its lipolytic fragment (AOD9604) on lipid metabolism following chronic treatment in obese mice and beta(3)-AR knock-out mice. Endocrinology. 2001;142:5182-5189.
  73. Fain JN, Ihle JH, Bahouth SW. Stimulation of lipolysis but not of leptin release by GH is abolished in adipose tissue from STAT5a and b knockout mice. Biochem Biophys Res Commun. 1999;263:201–205.
  74. Clodfelter KH, Holloway MG, Hodor P, Park SH, Ray WJ, Waxman DJ. Sex-dependent liver gene expression is extensive and largely dependent upon signal transducer and activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression of female genes revealed by microarray analysis. Mol Endocrinol. 2006;20:1333-1351.
  75. Sharma VM, Vestergaard ET, Jessen N, Kolind-Thomsen P, Nellemann B, Nielsen TS, Vendelbo MH, Møller N, Sharma R, Lee KY, Kopchick JJ, Jørgensen JOL, Puri V. Growth hormone acts along the PPARγ-FSP27 axis to stimulate lipolysis in human adipocytes. Am J Physiol Endocrinol Metab. 2019;316:E34-E42.
  76. Näntö-Salonen K, Muller HL, Hoffman AR, Vu TH, Rosenfeld RG. Mechanisms of thyroid hormone action on the insulin-like growth factor system: all thyroid hormone effects are not growth hormone mediated. Endocrinology. 1993;132:781-788.
  77. Veldhuis JD, Keenan DM, Bailey JN, Adeniji A, Miles JM, Paulo R, Cosma M, Soares-Welch C. Testosterone supplementation in older men restrains insulin-like growth factor's dose-dependent feedback inhibition of pulsatile growth hormone secretion. J Clin Endocrinol Metab. 2009;94:246-254.
  78. Colón E, Svechnikov KV, Carlsson-Skwirut C, Bang P, Soder O. Stimulation of steroidogenesis in immature rat Leydig cells evoked by interleukin-1alpha is potentiated by growth hormone and insulin-like growth factors. Endocrinology. 2005;146:221-30.
  79. Giudice LC. Insulin-like growth factor family in Graafian follicle development and function. J Soc Gynecol Investig. 2001;8(1 Suppl Proceedings):S26-9.
  80. Conover CA, Faessen GF, Ilg KE, Chandrasekher YA, Christiansen M, Overgaard MT, Oxvig C, Giudice LC. Pregnancy-associated plasma protein-a is the insulin-like growth factor binding protein-4 protease secreted by human ovarian granulosa cells and is a marker of dominant follicle selection and the corpus luteum. Endocrinology. 2001;142:2155-2161.
  81. Chrysis D, Zaman F, Chagin AS, Takigawa M, Sävendahl L. Dexamethasone induces apoptosis in proliferative chondrocytes through activation of caspases and suppression of the Akt-phosphatidylinositol 3'-kinase signaling pathway. Endocrinology. 2005;146:1391-1397.
  82. Bang P, Degerblad M, Thorén M, Schwander J, Blum W, Hall K. Insulin-like growth factor (IGF) I and II and IGF binding protein (IGFBP) 1, 2 and 3 in serum from patients with Cushing's syndrome. Acta Endocrinol (Copenh). 1993;128:397-404.
  83. Öberg D, Salemyr J, Örtqvist E, Juul A, Bang P. A longitudinal study of serum insulin-like growth factor-I levels over 6 years in a large cohort of children and adolescents with type 1 diabetes mellitus: A marker reflecting diabetic retinopathy. Pediatr Diabetes. 2018;19:972-978.
  84. Maile LA, Busby WH, Nichols TC, Bellinger DA, Merricks EP, Rowland M, Veluvolu U, Clemmons DR. A monoclonal antibody against alphaVbeta3 integrin inhibits development of atherosclerotic lesions in diabetic pigs. Sci Transl Med. 2010;2:18ra11.
  85. Vella A, Bouatia-Naji N, Heude B, Cooper JD, Lowe CE, Petry C, Ring SM, Dunger DB, Todd JA, Ong KK. Association analysis of the IGF1 gene with childhood growth, IGF-1 concentrations and type 1 diabetes. Diabetologia. 2008;51:811-815.
  86. Hedman CA, Frystyk J, Lindström T, Oskarsson P, Arnqvist HJ. Intraperitoneal insulin delivery to patients with type 1 diabetes results in higher serum IGF-I bioactivity than continuous subcutaneous insulin infusion. Clin Endocrinol (Oxf). 2014;81:58-62.
  87. Saukkonen T, Shojaee-Moradie F, Williams RM, Amin R, Yuen KC, Watts A, Acerini CL, Umpleby AM, Dunger DB. Effects of recombinant human IGF-I/IGF-binding protein-3 complex on glucose and glycerol metabolism in type 1 diabetes. Diabetes. 2006;55:2365-2370.
  88. Acerini CL, Patton CM, Savage MO, Kernell A, Westphal O, Dunger DB. Randomised placebo-controlled trial of human recombinant insulin-like growth factor I plus intensive insulin therapy in adolescents with insulin-dependent diabetes mellitus. Lancet. 1997;350:1199-1204.
  89. Woods KA, Camacho-Hübner C, Savage MO, Clark AJ. Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. N Engl J Med. 1996;335:1363-1367.
  90. Woods KA, Camacho-Hübner C, Bergman RN, Barter D, Clark AJ, Savage MO. Effects of insulin-like growth factor I (IGF-I) therapy on body composition and insulin resistance in IGF-I gene deletion. J Clin Endocrinol Metab. 2000;85:1407-1411.
  91. Dohm GL, Elton CW, Raju MS, Mooney ND, DiMarchi R, Pories WJ, Flickinger EG, Atkinson SM Jr, Caro JF. IGF-I--stimulated glucose transport in human skeletal muscle and IGF-I resistance in obesity and NIDDM. Diabetes. 1990;39:1028-1032.
  92. Nakae J, Kato M, Murashita M, Shinohara N, Tajima T, Fujieda K. Long-term effect of recombinant human insulin-like growth factor I on metabolic and growth control in a patient with leprechaunism. J Clin Endocrinol Metab. 1998;83:542-549.
  93. Dehghani SM, Karamifar H, Hamzavi SS, Haghighat M, Malek-Hosseini SA. Serum insulinlike growth factor-1 and its binding protein-3 levels in children with cirrhosis waiting for a liver transplant. Exp Clin Transplant. 2012;10:252-257.
  94. Moayeri H, Oloomi Z. Prevalence of growth and puberty failure with respect to growth hormone and gonadotropins secretion in beta-thalassemia major. Arch Iran Med. 2006;9:329-334.
  95. Alisi A, Pampanini V, De Stefanis C, Panera N, Deodati A, Nobili V, Cianfarani S. Expression of insulin-like growth factor I and its receptor in the liver of children with biopsy-proven NAFLD. PLoS One. 2018;13:e0201566.
  96. E CY, Cai Y, Sun BZ, Guan LY, Jiang T. Hepatic insulin-like growth factor receptor is upregulated by activation of the GSK3B-FOXO3 pathway after partial hepatectomy. J Biol Regul Homeost Agents. 2017;31:549-555.
  97. Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J, Pratt KL, Bezrodnik L, Jasper H, Tepper A, Heinrich JJ, Rosenfeld RG. Growth hormone insensitivity associated with a STAT5b mutation. N Engl J Med. 2003;349:1139-1147.
  98. Binder G, Neuer K, Ranke MB, Wittekindt NE. PTPN11 mutations are associated with mild growth hormone resistance in individuals with Noonan syndrome. J Clin Endocrinol Metab. 2005;90:5377-81.
  99. De Rocca Serra-Nédélec A, Edouard T, Tréguer K, Tajan M, Araki T, Dance M, Mus M, Montagner A, Tauber M, Salles JP, Valet P, Neel BG, Raynal P, Yart A. Noonan syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1 release via growth hormone-induced ERK hyperactivation, which contributes to short stature. Proc Natl Acad Sci U S A. 2012;109:4257-4262.
  100. Vesterlund M, Zadjali F, Persson T, Nielsen ML, Kessler BM, Norstedt G, Flores-Morales A. The SOCS2 ubiquitin ligase complex regulates growth hormone receptor levels. PLoS One. 2011;6:e25358.
  101. Hoppe C, Mølgaard C, Juul A, Michaelsen KF. High intakes of skimmed milk, but not meat, increase serum IGF-I and IGFBP-3 in eight-year-old boys. Eur J Clin Nutr. 2004;58):1211-1216.
  102. Hoppe C, Mølgaard C, Vaag A, Barkholt V, Michaelsen KF. High intakes of milk, but not meat, increase s-insulin and insulin resistance in 8-year-old boys. Eur J Clin Nutr. 2005;59:393-398.
  103. Mogami H, Yura S, Itoh H, Kawamura M, Fujii T, Suzuki A, Aoe S, Ogawa Y, Sagawa N, Konishi I, Fujii S. Isocaloric high-protein diet as well as branched-chain amino acids supplemented diet partially alleviates adverse consequences of maternal undernutrition on fetal growth. Growth Horm IGF Res. 2009;19:478-485.
  104. D'Esposito V, Passaretti F, Hammarstedt A, Liguoro D, Terracciano D, Molea G, Canta L, Miele C, Smith U, Beguinot F, Formisano P. Adipocyte-released insulin-like growth factor-1 is regulated by glucose and fatty acids and controls breast cancer cell growth in vitro. Diabetologia. 2012;55:2811-2822.
  105. Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586-623.
  106. DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell. 1991;64:849-859.
  107. Rossignol S, Netchine I, Le Bouc Y, Gicquel C. Epigenetics in Silver-Russell syndrome. Best Pract Res Clin Endocrinol Metab. 2008;22:403-414.
  108. Clemmons DR. Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol. 2018;61:T139-T169.
  109. Banskota NK, Carpentier JL, King GL. Processing and release of insulin and insulin-like growth factor I by macro- and microvascular endothelial cells. Endocrinology. 1986;119:1904-1913.
  110. Maile LA, Badley-Clarke J, Clemmons DR. The association between integrin-associated protein and SHPS-1 regulates insulin-like growth factor-I receptor signaling in vascular smooth muscle cells. Mol Biol Cell. 2003;14:3519-3528.
  111. Bang P, Ahlsén M, Berg U, Carlsson-Skwirut C. Free insulin-like growth factor I: are we hunting a ghost? Horm Res. 2001;55 Suppl 2:84-93.
  112. Bang P, Thorell A, Carlsson-Skwirut C, Ljungqvist O, Brismar K, Nygren J. Free dissociable IGF-I: Association with changes in IGFBP-3 proteolysis and insulin sensitivity after surgery. Clin Nutr. 2016;35:408-413.
  113. Juul A, Holm K, Kastrup KW, Pedersen SA, Michaelsen KF, Scheike T, Rasmussen S, Müller J, Skakkebaek NE. Free insulin-like growth factor I serum levels in 1430 healthy children and adults, and its diagnostic value in patients suspected of growth hormone deficiency. J Clin Endocrinol Metab. 1997;82:2497-2502.
  114. Ekström K, Pulkkinen MA, Carlsson-Skwirut C, Brorsson AL, Ma Z, Frystyk J, Bang P. Tissue IGF-I Measured by Microdialysis Reflects Body Glucose Utilization After rhIGF-I Injection in Type 1 Diabetes.J Clin Endocrinol Metab. 2015;100:4299-4306.
  115. Baxter RC. Insulin-like growth factor binding proteins in the human circulation: a review. Horm Res. 1994;42:140-144.
  116. Bang P, Stangenberg M, Westgren M, Rosenfeld RG. Decreased ternary complex formation and predominance of a 29 kDa IGFBP-3 fragment in human fetal serum. Growth Regul. 1994;4:68-76.
  117. Ekström K, Carlsson-Skwirut C, Ritzén EM, Bang P. Insulin-like growth factor-I and insulin-like growth factor binding protein-3 cotreatment versus insulin-like growth factor-I alone in two brothers with growth hormone insensitivity syndrome: effects on insulin sensitivity, body composition and linear growth. Horm Res Paediatr. 2011;76:355-366.
  118. Giudice LC, Farrell EM, Pham H, Lamson G, Rosenfeld RG. Insulin-like growth factor binding proteins in maternal serum throughout gestation and in the puerperium: effects of a pregnancy-associated serum protease activity. J Clin Endocrinol Metab. 1990;71:806-816.
  119. Hossenlopp P, Segovia B, Lassarre C, Roghani M, Bredon M, Binoux M. Evidence of enzymatic degradation of insulin-like growth factor-binding proteins in the 150K complex during pregnancy. J Clin Endocrinol Metab. 1990;71:797-805.
  120. Diamandi A, Mistry J, Krishna RG, Khosravi J. Immunoassay of insulin-like growth factor-binding protein-3 (IGFBP-3): new means to quantifying IGFBP-3 proteolysis. J Clin Endocrinol Metab. 2000;85:2327-2333.
  121. Ahlsén M, Carlsson-Skwirut C, Jonsson AP, Cederlund E, Bergman T, Bang P. A 30-kDa fragment of insulin-like growth factor (IGF) binding protein-3 in human pregnancy serum with strongly reduced IGF-I binding. Cell Mol Life Sci. 2007;64:1870-1880.
  122. Yan X, Payet LD, Baxter RC, Firth SM. Activity of human pregnancy insulin-like growth factor binding protein-3: determination by reconstituting recombinant complexes. Endocrinology. 2009;150:4968-4976.
  123. Zachrisson I, Brismar K, Carlsson-Skwirut C, Dahlquist G, Wallensteen M, Bang P. Increased 24 h mean insulin-like growth factor binding protein-3 proteolytic activity in pubertal type 1 diabetic boys. Growth Horm IGF Res. 2000;10:324-331.
  124. Bang P, Brismar K, Rosenfeld RG. Increased proteolysis of insulin-like growth factor-binding protein-3 (IGFBP-3) in noninsulin-dependent diabetes mellitus serum, with elevation of a 29-kilodalton (kDa) glycosylated IGFBP-3 fragment contained in the approximately 130- to 150-kDa ternary complex. J Clin Endocrinol Metab. 1994;78:1119-1127.
  125. Bang P, Fielder PJ. Human pregnancy serum contains at least two distinct proteolytic activities with the ability to degrade insulin-like growth factor binding protein-3. Endocrinology. 1997;138:3912-3917.
  126. Rajah R, Katz L, Nunn S, Solberg P, Beers T, Cohen P. Insulin-like growth factor binding protein (IGFBP) proteases: functional regulators of cell growth. Prog Growth Factor Res. 1995;6:273-84.
  127. Martin DC, Fowlkes JL, Babic B, Khokha R. Insulin-like growth factor II signaling in neoplastic proliferation is blocked by transgenic expression of the metalloproteinase inhibitor TIMP-1. J Cell Biol. 1999;146:881-892.
  128. Gaidamauskas E, Gyrup C, Boldt HB, Schack VR, Overgaard MT, Laursen LS, Oxvig C.IGF dependent modulation of IGF binding protein (IGFBP) proteolysis by pregnancy-associated plasma protein-A (PAPP-A): multiple PAPP-A-IGFBP interaction sites. Biochim Biophys Acta. 2013 Mar;1830(3):2701-9. doi: 10.1016/j.bbagen.2012.11.002.PMID: 23671931
  129. Bang P, Ahmed SF, Argente J, Backeljauw P, Bettendorf M, Bona G, Coutant R, Rosenfeld RG, Walenkamp MJ, Savage MO. Identification and management of poor response to growth-promoting therapy in children with short stature. Clin Endocrinol (Oxf). 2012;77:169-181.
  130. Bang P, Polak M, Woelfle J, Houchard A; EU IGFD Registry Study Group. Effectiveness and Safety of rhIGF-1 Therapy in Children: The European Increlex® Growth Forum Database Experience. Horm Res Paediatr. 2015;83:345-357.
  131. Murray and Clayton. Disorders of growth hormone in childhood. MDText.com 2022
  132. Cabrera-Salcedo C, Mizuno T, Tyzinski L, Andrew M, Vinks AA, Frystyk J, Wasserman H, Gordon CM, Hwa V, Backeljauw P, Dauber A. Pharmacokinetics of IGF-1 in PAPP-A2-Deficient Patients, Growth Response, and Effects on Glucose and Bone Density. J Clin Endocrinol Metab. 2017 Dec 1;102(12):4568-4577. doi: 10.1210/jc.2017-01411.
  133. Muthuvel G, Dauber A, Alexandrou E, Tyzinski L, Andrew M, Hwa V, Backeljauw P. Five-Year Therapy with Recombinant Human Insulin-Like Growth Factor-1 in a Patient with PAPP-A2 Deficiency. Horm Res Paediatr. 2023;96(5):449-457. doi: 10.1159/000529071. Epub 2023 Jan 16.
  134. Muñoz-Calvo MT, Barrios V, Pozo J, Chowen JA, Martos-Moreno GÁ, Hawkins F, Dauber A, Domené HM, Yakar S, Rosenfeld RG, Pérez-Jurado LA, Oxvig C, Frystyk J, Argente J. Treatment With Recombinant Human Insulin-Like Growth Factor-1 Improves Growth in Patients With PAPP-A2 Deficiency. J Clin Endocrinol Metab. 2016 Nov;101(11):3879-3883. doi: 10.1210/jc.2016-2751. Epub 2016 Sep 20.

Evaluation and Treatment of Dyslipidemia in the Elderly

ABSTRACT

 

The definition of elderly is arbitrary. In this chapter we will define elderly as greater than 75 years of age because both the US and European lipids guidelines use this age to differentiate therapy recommendations. Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in the elderly. Age is a key risk factor for ASCVD and with identical risk factors the 10-year risk of an ASCVD event markedly increases with age. In fact, an older individual with excellent risk factors can still have a high risk for having an ASCVD event. ASCVD begins early in life and progresses until it leads to clinical events later in life. The age that one develops clinical manifestations of ASCVD is dependent on the severity of individual risk factors, the number of risk factors, and the duration of exposure to the risk factors. Elderly individuals have a long exposure to risk factors so even when the risk factors are relatively modest the cumulative effects can be sufficient to result in clinical ASCVD events. This explains why age is such a key variable in determining the risk of developing an ASCVD event. Cardiovascular outcome studies have demonstrated that lowering LDL-C levels with statins, ezetimibe, or PCSK 9 monoclonal antibodies will reduce ASCVD events in elderly patients with pre-existing cardiovascular disease (secondary prevention). In elderly patients without cardiovascular disease (primary prevention) the available data does not definitively demonstrate a decrease in ASCVD events with statin or ezetimibe therapy but is suggestive of a benefit (note there are no primary prevention trials with PCSK9 inhibitors). Additional data is required to determine if bempedoic acid and icosapent ethyl reduce ASCVD events in patients ≥ 75 years of age. Studies are currently underway to provide definitive information on whether statin therapy is beneficial as primary prevention in the elderly. In deciding whether to treat an elderly patient with lipid lowering drugs one needs to consider the following factors; the higher the LDL-C level the greater the benefit of lowering LDL-C, the greater the decrease in LDL-C the greater the benefit, the higher the absolute risk of ASCVD the greater the benefit of lowering LDL-C, life expectancy, competing non-cardiovascular disorders, risk of drug side effects, potential for drug interactions, and patient preferences. In elderly patients without pre-existing ASCVD one should estimate the patient’s risk of developing ASCVD events and in conjunction with the general principles described above discuss with the patient a treatment plan. Determining the coronary calcium score can be helpful if there is uncertainty regarding the appropriate decision. If the decision is to treat our goal in primary prevention patients is often an LDL-C < 100mg/dL but in high-risk patients our goal may be an LDL-C < 70mg/dL. Elderly patients with ASCVD should be treated with lipid lowering drugs to reduce ASCVD unless there are contraindications. At a minimum our goal is an LDL-C < 70mg/dL but we would prefer an LDL-C < 55mg/dL if they can be achieved with a statin + ezetimibe. In very high-risk patients our goal is an LDL-C < 55mg/dL and adding a PCSK9 inhibitor may be required to achieve these levels in some patients. Age per se should not be used to withhold therapy with lipid lowering drugs that can reduce the risk of ASCVD events.

 

INTRODUCTION

 

Due to a decreasing birth rate and a longer life expectancy the population is getting older. According to the US census in 2020 there were approximately 50 million people between 65 and 84 years of age (14.9% of the total population) and approximately 6 million between 85 and 99 years of age (1.89% of the total population). The number of Americans ages 65 and older is projected to increase to 82 million by 2050 (23% of the total population). World-wide there are 703 million people aged 65 or older, which is projected to reach 1.5 billion by 2050 (1 in 6 people). It is well recognized that atherosclerotic cardiovascular disease (ASCVD) increases with age and is a major cause of morbidity and mortality in the elderly. In addition to an increased risk of coronary artery disease there is more than a doubling of the prevalence of peripheral arterial disease, cerebrovascular disease, and abdominal aortic aneurism with each decade of life (1). Unfortunately, the elderly (≥ 75 years of age) have not been well represented in lipid lowering cardiovascular outcome trials.

 

The definition of elderly is arbitrary. In this chapter we will define elderly as greater than 75 years of age because both the US and European guidelines use this age to differentiate therapy recommendations. In both the “Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines” and the “2019 ESC/EAS Guidelines for the Management of Dyslipidemias: Lipid Modification to Reduce Cardiovascular Risk” the recommendations for those over 75 years of age differ from recommendations for younger individuals (2,3). Thus, where possible we will focus on studies in individuals greater than 75 years of age.

 

LIPID LEVELS IN THE ELDERLY

 

Lipid levels in US adults from the National Health and Nutrition Examination Survey (NHANES) 2003-2004 are shown in Table 1 (4). Compared to 30–69-year-olds there is a slight decrease in LDL-C, non-HDL-C, and triglycerides with similar HDL-C levels in individuals 70-79 years of age. Other cross-sectional studies have reported similar results (5-11). Prospective studies with longitudinal follow-up have also observed small decreases in total cholesterol, LDL-C, and HDL-C levels in men and women as they become elderly (6,7,9,12-14). It should be noted that the changes in lipid levels reported with aging are relatively small and vary somewhat from study to study. The clinical significance of these small changes is uncertain.

 

Table 1. Lipid Levels in U.S. Adults (NHANES 2003-2004)

Age

LDL-C (mg/dL)

Non-HDL-C (mg/dL)

HDL-C (mg/dL)

Triglycerides (mg/dL)

20-29

104

126

54

105

30-39

120

146

54

118

40-49

124

152

53

144

50-59

123

154

55

141

60-69

126

157

54

145

70-79

119

148

56

133

 

Studies have demonstrated that older individuals have an exaggerated postprandial lipemia compared with younger individuals (15,16). While elevated postprandial triglycerides is associated with an increased risk of ASCVD whether this plays a causal role in increasing ASCVD is uncertain.

 

It is well recognized that with increasing age the likelihood of other medical disorders increases and this can affect lipid levels. For example, inflammation and infections can decrease LDL-C and HDL-C levels (17). Additionally, poor nutrition due to illness or social-economic factors could decrease lipid levels in the elderly. Finally, frailty is a syndrome associated with aging and increases with age. It is usually associated with a lowering of total, LDL-C, and non-HDL cholesterol levels (18-20).

 

AGE IS AN IMPORTANT RISK FOR ATHEROSCLEROTIC CARDIOVASCULAR DISEASE

 

The clearest way to illustrate the importance of age as a key risk factor for atherosclerotic cardiovascular disease (ASCVD) is to compare the 10-year risk at different ages using an updated version of the AHA/ACC pooled cohort equation. Shown in table 2 are four examples of the effect of age on 10-year risk in different clinical situations that demonstrate the marked effect of age on ASCVD risk. Similarly, using the SCORE risk calculator for determining the 10-year risk of fatal cardiovascular disease also demonstrates the very large impact of age on risk (figure 1). It is obvious that age is a major determinant of ASCVD risk.

 

Table 2. Ten Year Risk of Developing ASCVD

 

Age 55

Age 65

Age 75

Male, white, BP 130, TC 200, HDL-C 45, non-smoker, no diabetes

6.3%

13.9%

26.2%

Female, African American, BP120, TC 180, HDL-C 50, non-smoker, no diabetes

2.8%

6.3%

13.2%

Male, African American, BP140, TC 200, HDL-C 50, smoker, no diabetes

10.1%

14.9%

20.5%

Female, white, BP 140, TC 180, HDL-C 50, non-smoker, diabetes

4.4%

12.2%

32.9%

https://tools.acc.org/ascvd-risk-estimator-plus/#!/calculate/estimate/.

BP= systolic BP mm Hg, TC= total cholesterol mg/dL.

 

Figure 1. Systematic Coronary Risk Estimation chart for European populations at high cardiovascular disease risk (from 2019 ESC/EAS Guidelines for the management of dyslipidaemias (3)).

 

In fact, an older individual with excellent risk factors can have a high risk for having an ASCVD event. For example, using the AHA/ACC pooled cohort equation a 75-year-old white male with a total cholesterol of 180mg/dL, an HDL-C of 50mg/dL, a blood pressure of 120/80 mmHg, who is not diabetic, doesn’t smoke, and is on no medications still has a 10-year risk for an ASCVD event of 21.7%. A 75-year-old white female with the same risk factors also has a relatively high 10-year risk (14.1%). Using the SCORE estimator (figure 1) for a fatal CVD event it is also apparent that many older individuals, particularly males, are at high risk even when they are non-smokers with an excellent total cholesterol and blood pressure. For example, a 70-year-old male, non-smoker with a total cholesterol of 160mg/dL and systolic BP of 120 mmHg still has a 13% 10-year risk of death from CVD.

 

WHY ARE OLDER INDIVIDUALS AT HIGHER RISK FOR ASCVD?

 

It is widely recognized that atherosclerosis begins early in life and slowly progresses ultimately resulting in clinical manifestations later in life (21). Numerous studies have demonstrated the presence of atherosclerosis in young individuals (22-27). In the Bogalusa Heart Study autopsies were performed on 204 young people 2 to 39 years of age (22,28). In the coronary arteries fatty streaks were very common (50 percent at 2 to 15 years of age and 85 percent at 21 to 39 years of age). More advanced raised fibrous-plaque lesions in the coronary arteries were present in 8 percent of individuals 2 to 15 years of age and 69 percent of individuals 26 to 39 years of age. The extent of the atherosclerotic lesions correlated positively with BMI, systolic and diastolic BP, total cholesterol, LDL-C, and triglyceride levels and negatively with HDL-C levels. The extent of the atherosclerotic lesions was greatest in individuals who had multiple risk factors. The Pathobiological Determinants of Atherosclerosis in Youth [PDAY] study examined the effect of risk factors for atherosclerosis in 1079 men and 364 women 15 through 34 years of age who died due to accidents, homicide, or suicide (23,29). Atherosclerosis of the aorta and right coronary artery was measured and increased with age, LDL-C levels, glycohemoglobin levels, BMI, and smoking while HDL-C levels were negatively associated with the extent of fatty streaks and raised lesions in the aorta and right coronary artery. Finally, in a study of US service members (mean age 25.9 years; range 18-59 years; 98.3% male) who died of combat or unintentional injuries (n= 3832) the effect of risk factors on coronary atherosclerosis was determined (27). Atherosclerosis prevalence was increased by age, dyslipidemia, hypertension, and obesity. Taken together these studies clearly demonstrate that atherosclerosis begins early in life with the prevalence increasing with age and the extent and onset of lesions is influenced by risk factors, including dyslipidemia.

 

Moreover, the presence of risk factors early in life is associated with an increase in atherosclerosis later in life (30-32). A meta-analysis that included 4380 participants from 4 prospective studies that collected cardiovascular risk factor data during childhood (age 3 to 18 years) and measured carotid intima-media thickness (CIMT) in adulthood (age 20 to 45 years) reported that total cholesterol, triglycerides, BP, and BMI measured in childhood were predictive of elevated CIMT in adults (33). Additionally, increased LDL-C and/or decreased HDL-C during adolescence predict an increase in CIMT later in life (34). Importantly, an increased total cholesterol or BP early in life also predicted an increased risk of developing cardiovascular disease later in life (35-38).

 

Genetic studies have further illustrated the key role of risk factors and duration of exposure to the risk factor as key variables determining the time when clinical manifestations of ASCVD occur. In patients with homozygous familial hypercholesterolemia (FH) LDL-C are markedly elevated and cardiovascular events can occur early in life. Greater than 50% of untreated patients with homozygous FH develop clinically significant ASCVD by the age of 30 and cardiovascular events can occur before age 10 in some patients (39). In patients with heterozygous FH LDL-C levels are elevated but not to the levels seen with homozygous FH and cardiovascular events occur later in life but still at a relatively younger age. Untreated males with heterozygous FH have a 50% risk for a fatal or non-fatal myocardial infarction by 50 years of age whereas untreated females have a 30% chance by age 60 (39). Conversely, individuals with genetic variants in PCSK9, HMG-CoA reductase, LDL receptor, NPC1L1, or ATP citrate lyase that lead to a decrease in LDL-C levels have a reduced risk of developing cardiovascular events (40,41). The relationship between genetic disorders that alter LDL-C levels and the time to develop clinical cardiovascular events is illustrated in figure 2. The figure clearly illustrates that the age when one clinically manifests ASCVD depends on the level of LDL-C. With very high LDL-C levels clinical events occur early in life and with low LDL-C levels events will occur at an older age leading to the concept of LDL years.

 

Figure 2. Relationship between cumulative LDL-C exposure and age of developing cardiovascular disease. (from (41)).

 

The degree and duration of other risk factors also seems to play a role in when the clinical manifestations of ASCVD are expressed. For example, for cigarette smoking, cigarettes/day, smoking duration, and pack-years all increase the risk of cardiovascular disease (42). Interestingly smoking fewer cigarettes/day for a longer duration was more deleterious than smoking more cigarettes/day for a shorter duration (42,43). Additionally, while smoking cessation lowers the risk of ASCVD events an increased risk persists for decades after smoking cessation (44). These observations suggest that the effect of smoking is related to the number of cigarettes smoked and the duration of the smoking (i.e., pack years). Similarly, in patients with diabetes glycemic control and duration of diabetes influences the development of ASCVD complications (45-48). At any given age, a 10-year longer diabetes duration was associated with a 1.1-1.5-fold increased risk of stroke and 1.5-2.0-fold increased risk of MI (45).

 

Thus, ASCVD begins early in life and progresses until it leads to clinical events such as a myocardial infarction or stroke later in life. The age that one develops clinical manifestations of ASCVD is dependent on the severity of individual risk factors, the number of risk factors, and the duration of exposure to the risk factors. Elderly individuals have a long exposure to risk factors so even when the risk factors are relatively modest the cumulative effects can be sufficient to result in clinical ASCVD events. This explains why age is such a key variable in determining the risk of developing an ASCVD event.

 

DOES LIPID LOWERING REDUCE EVENTS IN THE ELDERLY

 

Below we discuss lipid lowering drug studies that report the effect on cardiovascular outcomes that are relevant to clinical decisions in elderly individuals. For additional and more detailed information on lipid lowering cardiovascular outcome studies see the Endotext chapters on “Cholesterol Lowering Drugs” and “Triglyceride Lowering Drugs” (49,50).

 

Statins

 

Few statin studies have focused on lowering LDL-C in elderly patients, which we define as individuals greater than 75 years of age. The Prosper Trial determined the effect of pravastatin 40mg/day (n= 2891) vs. placebo (n= 2913) on cardiovascular events in older subjects (70-82 years of age) with pre-existing vascular disease or who were at high risk for vascular disease (51). The average age in this trial was 75 years of age and approximately 45% had cardiovascular disease. As expected, pravastatin treatment lowered LDL-C by 34% compared to the placebo group. The primary end point was coronary death, non-fatal myocardial infarction, and fatal or non-fatal stroke, which was reduced by 15% (HR 0.85, 95% CI 0.74-0.97, p=0.014). However, in the individuals without pre-existing cardiovascular disease pravastatin did not significantly reduce ASCVD events (HR 0.94; CI 0.77–1.15). In contrast, in individuals with cardiovascular disease pravastatin therapy significantly reduced ASCVD events (HR 0.78, CI 0.66–0.93). Thus, this study demonstrated benefits of statin therapy in the elderly with cardiovascular disease but failed to demonstrate benefit in the elderly without cardiovascular disease.

 

A meta-analysis by the Cholesterol Treatment Trialists of 28 trials with 14,483 of 186,854 found a reduction in LDL-C levels with statin therapy that was similar in the participants ≥75 years of age compared to younger individuals. Moreover, statin therapy resulted in a decrease in cardiovascular events in all age groups including participants ≥75 years of age (Figure 3) (52). In the participants ≥75 there was a 13% reduction in ASCVD events per 39mg/dL decrease in LDL-C (RR 0.87; 95% CI 0.77–0.99). This analysis included four trials done exclusively among people with heart failure or receiving renal dialysis, for whom statin therapy shows little or no benefit (50). A second analysis was performed after elimination of these four trials and there was an 18% reduction in ASCVD events per 39mg/dL decrease in LDL-C (RR 0.82; 95%CI 0.70-0.95). Similar to the Prosper Trial a decrease in ASCVD events was clearly demonstrated in individuals with pre-existing cardiovascular disease (secondary prevention) but in individuals without pre-existing cardiovascular disease (primary prevention) the decrease in ASCVD events was not statistically significant (Figure 4- analysis included all studies). After excluding the trials in patients with heart failure or receiving renal dialysis, statin therapy reduced major ASCVD events by 26% per 39mg/dL decrease in LDL-C (RR 0.74; 95% CI0.60 − 0.91) in patients with pre-existing cardiovascular disease but only by 8% in patients without pre-existing cardiovascular disease (RR 0.92; 95%CI 0.72 − 1.16).

 

Figure 3. Effect of Statin Treatment on Major Vascular Events. Modified from (44).

Figure 4. Effect of Statin Treatment on Major Vascular Events in Individuals With and Without Pre-Existing Cardiovascular Disease. Modified from (44).

 

A statin trial not included in the Cholesterol Treatment Trialists meta-analysis was carried out in patients with an ischemic stroke or a transient ischemic attack who were treated with statins and/or ezetimibe with a target LDL-C level < than 70mg/dL (n= 1430) or an LDL-C 90mg/dL to 110mg/dL (n= 1430) (53). The primary end point was ischemic stroke, myocardial infarction, new symptoms leading to urgent coronary or carotid revascularization, or death from cardiovascular causes. The mean LDL-C level was 65mg/dL in the lower-target group and 96 mg/dL in the higher-target group. After median of 3.5 years the primary end point occurred in 8.5% of the patients in the lower-target group and 10.9% of the patients in the higher target group (HR 0.78; 95% CI 0.61 to 0.98; P=0.04). In patients < 65 years of age only a 7% decrease in the primary end point was observed (HR 0.93; 95%CI 0.63–1.36) whereas more impressive decreases in the primary endpoint were observed in patients 65-75 years of age (37% decrease; HR 0.63 95% CI 0.42–0.95) and > 75 years of age (23% decrease; HR 0.77; 95%CI 0.49–1.22). These results are consistent with the Cholesterol Treatment Trialists meta-analysis demonstrating that elderly patients with pre-existing cardiovascular disease lowering LDL-C levels reduces ASCVD events.

 

There are observational studies demonstrating that statin treatment for the primary prevention of ASCVD is effective in older patients (54-59). For example, in US veterans ≥75 years of age and free of ASCVD at baseline, new statin use was significantly associated with a lower risk of ASCVD events (HR 0.92; 95% CI 0.91-0.94) and cardiovascular mortality (HR 0.80; 95% CI 0.78-0.81) when compared to statin nonusers (55). Similarly, in a Danish nationwide cohort initiation of statin therapy in patients > 70 years of age without pre-existing cardiovascular disease there was a 23% lower risk of major vascular events per 39mg/dL decrease in LDL-C (HR 0.77; 95% CI 0.71-0.83), which was similar to what was observed in younger individuals (54). Finally, in nursing home residents without ASCVD statin use reduced all-cause mortality in individuals with and without dementia (59). These observational studies while suggestive of a benefit of statin therapy for primary prevention in older individuals cannot provide definitive proof as there is always the possibility of residual confounding. Nevertheless, they provide additional support that statin therapy provides benefits in elderly patients without pre-existing cardiovascular disease.

 

Thus, in older patients with cardiovascular disease lowering LDL-C levels with statins clearly reduces cardiovascular events but in older patients without cardiovascular disease the data demonstrating that statins reduce cardiovascular events is less robust but suggests a reduction in ASCVD events.

 

Ezetimibe

 

IMPROVE-IT TRIAL

 

The IMPROVE-IT Trial tested whether the addition of ezetimibe to statin therapy would provide an additional beneficial effect in patients with the acute coronary syndrome (60). The IMPROVE-IT Trial was a large trial with over 18,000 patients randomized to simvastatin 40mg vs. simvastatin 40mg + ezetimibe 10mg per day. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 54mg/dL in the statin + ezetimibe group. There was a small but significant 6.4% decrease in major cardiovascular events (cardiovascular death, MI, documented unstable angina requiring rehospitalization, coronary revascularization, or stroke) in the statin + ezetimibe group (HR 0.936; 95% CI 0.887-0.988; p=0.016). Cardiovascular death, non-fatal MI, or non-fatal stroke were reduced by 10% (HR 0.90; 95% CI 0.84-0.97; p=0.003). The effect of age on the benefits of statin + ezetimibe therapy is shown in figure 5. In elderly individuals (≥ 75 years of age) the combination of ezetimibe and simvastatin reduced ASCVD events.

 

Figure 5. Primary endpoint in the IMPROVE-IT trial in different age groups. Modified from (60).

 

EWTOPIA 75 TRIAL

 

EWTOPIA 75 was a multicenter, randomized trial in Japan that examined the preventive efficacy of ezetimibe for patients aged ≥75 years (mean age 80.6 years), with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease (primary prevention) who were not taking lipid lowering drugs (61). Patients were randomized to ezetimibe 10mg (n=1,716) or usual care (n=1,695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, myocardial infarction, coronary revascularization, or stroke. In the ezetimibe group LDL-C was decreased by 25.9% and non-HDL-C by 23.1% while in the usual care group LDL-C was decreased by 18.5% and non-HDL-C by 16.5% (p<0.001 for both lipid parameters). By the end of the trial 9.6% of the patients in the usual care group and 2.1% of the ezetimibe group were taking statins. Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; P=0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; P=0.039) and coronary revascularization by 62% (HR 0.38; P=0.007) in the ezetimibe group vs. the control group. There was no difference in the incidence of stroke or all-cause mortality between the groups. It should be noted that the reduction in cardiovascular events was much greater than one would expect based on the absolute difference in LDL-C levels (121mg/dL in ezetimibe group vs. 132mg/dL in usual care group). As stated by the authors “Given the open-label nature of the trial, its premature termination, and issues with follow-up, the magnitude of benefit observed should be interpreted with caution.” Nevertheless, this study suggests that lowering LDL-C in elderly individuals without cardiovascular disease can reduce ASCVD events.

 

RACING TRIAL

 

The RACING trial was a randomized, open-label trial in patients with ASCVD carried out in South Korea (62). Patients were randomly assigned to either rosuvastatin 10 mg with ezetimibe 10 mg (n= 1894) or rosuvastatin 20 mg (n= 1886). The primary endpoint was cardiovascular death, major cardiovascular events, or non-fatal stroke. The median LDL-C level during the study was 58mg/dL in the combination therapy group and 66mg/dL in the statin monotherapy group (p<0.0001). The primary endpoint occurred in 9.1% of the patients in the combination therapy group and 9.9% of the patients in the high-intensity statin monotherapy group (non-inferior). Non-inferiority was observed in patients with baseline LDL-C levels < 100mg/dL and >100mg/dL (63).

 

In the RACING trial 574 participants (15.2%) were aged ≥75 years and there was no difference in the primary endpoint between the combination therapy group and the high-intensity statin monotherapy group in these elderly participants (64). However, in participants ≥75 years of age moderate-intensity statin with ezetimibe combination therapy was associated with lower rates of drug related intolerance with drug discontinuation or dose reduction (2.3% vs 7.2%; P = 0.010).

 

This study demonstrates that moderate intensity statin plus ezetimibe was non-inferior to high-intensity statin therapy with regards to cardiovascular death, major cardiovascular events, or non-fatal stroke. The lower prevalence of discontinuation or dose reduction caused by intolerance to the study drug was seen with combination therapy indicating that using a moderate intensity dose of a statin plus ezetimibe is a useful strategy in patients that do not tolerate high intensity statin therapy or where there are concerns about statin toxicity with high doses.

 

PCSK9 Inhibitors

 

FOURIER TRIAL

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (65). The primary end point was cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, myocardial infarction, or stroke. The median duration of follow-up was 2.2 years. Baseline LDL-C levels were 92mg/dL and evolocumab resulted in a 59% decrease in LDL levels (LDL-C level on treatment approximately 30mg/dL). In this trial 6233 of the participants were > 69 years of age and the decrease in LDL was similar in participants > 69 years of age and younger individuals (66). A 14% reduction in the primary endpoint (HR 0.86; 95% CI 0.74–0.99) and a 18% reduction in the secondary endpoint (HR 0.82; 95% CI 0.69–0.98) was observed in the participants > 69 years of age, which was similar to the decreases seen in younger individuals (66). The effect of treatment with evolocumab on the primary and secondary endpoint in specific age groups is shown in table 3 (66). These results demonstrate that lowering LDL-C with a PCSK9 inhibitor decreases ASCVD events in elderly patients.

 

Table 3. Effect of Evolocumab Treatment on Cardiovascular Outcomes in Different Age Groups

 

< 65

65-75

>75

Primary Endpoint

HR 0.86; 95%CI 0.78–0.94

HR 0.86; 95%CI 0.76–0.97

HR 0.78; 95%CI 0.60–1.02

Secondary Endpoint

HR 0.79; 95%CI 0.69–0.90

HR 0.82; 95%CI 0.70–0.95

HR 0.78 95%CI 0.58–1.04

For the primary endpoint the P interaction for the three age groups = 0.84

For the secondary endpoint the P interaction for the three age groups = 0.94.  

 

ODYSSEY TRIAL

 

The ODYSSEY trial was a multicenter, randomized, double-blind, placebo-controlled trial involving 18,924 patients who had an acute coronary syndrome 1 to 12 months earlier, an LDL-C level of at least 70 mg/dL, a non-HDL-C level of at least 100 mg/dL, or an apolipoprotein B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (67). Patients were randomly assigned to receive alirocumab 75 mg every 2 weeks or matching placebo. The dose of alirocumab was adjusted to target an LDL-C level of 25 to 50 mg/dL. The primary end point was a composite of death from coronary heart disease, nonfatal myocardial infarction, fatal or nonfatal ischemic stroke, or unstable angina requiring hospitalization. In this trial 5084 (26.9%) individuals were ≥ 65 years of age, 1007 (5.3%) ≥ 75 years of age, and 42 (0.2%) ≥ 85 years of age (68). The baseline and decrease in LDL-C levels were similar in participants ≥65 years of age and those <65 years of age (LDL-C at baseline approximately 94mg/dL and after 4 months of treatment approximately 40mg/dL) (67,68). In the individuals ≥ 65 years of age there was a 22% decrease in the primary endpoint (HR 0.78; 95% CI 0.68–0.91) and in those < 65 years of age a 11% decrease (HR 0.89; 95% CI 0.80–1.00) (68). The secondary endpoint of all-cause death, myocardial infarction, or ischemic stroke was also reduced in the ≥ 65 participants (HR 0.78; 95% CI 0.68–0.90) and < 65 participants (HR 0.91; 0.82–1.02) (68). In participants ≥ 75 years of age the primary endpoint was reduced by 15% (HR 0.85; 95% CI 0.64–1.13) (68). When plotted as a continuous variable the relative benefit of alirocumab over placebo on the primary endpoint was consistent across the entire age range (figure 6).

 

Figure 6. Relative benefit of alirocumab at various ages. Modified from reference (68).

These two studies demonstrate that lowering LDL-C with a PCSK9 inhibitor decreases ASCVD events in elderly patients with pre-existing cardiovascular disease.

 

Bempedoic Acid

 

The CLEAR Outcome trial was a double-blind, randomized, placebo-controlled trial involving patients with cardiovascular disease or at high risk of cardiovascular disease who were unable or unwilling to take statins ("statin-intolerant" patients) (69). The patients were randomized to bempedoic acid 180 mg (n= 6992) or placebo (n= 6978) and the median duration of follow-up was 40.6 months. In this trial 44% of the participants were between ≥65 to < 75 years of age and 15% were ≥ 75 years of age. As expected, LDL-C levels were decreased by 21% in the bempedoic group compared to placebo (29mg/dL difference). The primary endpoint, death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization, was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI 0.79- 0.96; P = 0.004). The effect of age on the primary endpoint is shown in table 4.

 

Table 4. Effect of Bempedoic on Cardiovascular Outcomes in Different Age Groups

< 65

HR 0.87; 95% CI 0.74-1.02

≥65-<75

HR 0.83, 95% CI 0.72-0.96

≥ 75

HR 0.95, 95% CI 0.77-1.16

Interaction P value = 0.60

 

Niacin and Fibrates

 

Because of the robust effect of statins in lowering LDL-C levels and cardiovascular events recent trials of both niacin and fibrates have focused on the addition of these lipid lowering drugs to statin therapy. The AIM-HIGH trial was designed to determine if the addition of Niaspan, an extended-release form of niacin, to aggressive statin therapy would result in a further reduction in ASCVD events in patients with pre-existing cardiovascular disease (70) while the HPS 2 Thrive trial determined the effect of adding extended-release niacin (2000mg/day) combined with laropiprant, a prostaglandin D2 receptor antagonist, to statin therapy on ASCVD events in  patients with pre-existing vascular disease (71). Unfortunately, both of these trials failed to demonstrate a decrease in ASCVD events with the addition of niacin to statin therapy. The absence of benefit and increased side effects from niacin therapy has markedly reduced enthusiasm for treating patients with niacin to reduce ASCVD event. For additional details on these two studies and other niacin cardiovascular outcome studies see the Endotext chapter “Triglyceride Lowering Drugs” (49).

 

The ACCORD-LIPID Trial was designed to determine if the addition of fenofibrate to aggressive statin therapy in patients with pre-existing cardiovascular disease or at high risk for developing cardiovascular disease would result in a further reduction in cardiovascular disease in patients with Type 2 diabetes (72). The PROMINENT trial determined whether pemafibrate, a new selective PPAR-alpha activator, in patients on statin therapy with diabetes and pre-existing cardiovascular disease or at high risk for developing cardiovascular disease would reduce cardiovascular events (73). Disappointingly, neither trial demonstrated benefits from adding a fibrate to statin therapy. For additional details on these two studies and other fibrate ASCVD outcome studies see the Endotext chapter “Triglyceride Lowering Drugs” (49).

 

Thus, there is currently little enthusiasm for adding either niacin or a fibrate to statin therapy to reduce ASCVD events. One should recognize that like all studies these trials have limitations, that are discussed in detail in reference (49), and it is possible that future trials could resurrect the use of niacin and/or fibrates for decreasing ASCVD.

 

Omega-3-Fatty Acids (Fish Oil)

 

Numerous studies have determined the effect of low dose fish oil (< 1 gram per day) on ASCVD and found that they do not consistently reduce the risk of cardiovascular disease (49). Described below are ASCVD outcome studies that have used higher doses.

 

JAPAN EPA LIPID INTERVENTION STUDY (JELIS)

 

JELIS was an open label study without a placebo in patients with total cholesterol levels > 254mg/dL with cardiovascular disease (n= 3,664) or without cardiovascular disease (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9,326) or statin alone (n= 9,319) with a 5-year follow-up (74). The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal myocardial infarction, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. Total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TGs were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA plus statin group the primary endpoint occurred in 2.8% of the patients vs. 3.5% of the patients in the statin alone group (19% decrease; p = 0.011). In participants < 61 years of age the primary endpoint was reduced by 24% (HR 0.76; 95%CI 0.57–1.00) while in individuals ≥ 61 years of age the primary endpoint was reduced by 16% (HR 0.84; 95% CI 0.68–1.02; p interaction 0.57). Unstable angina and non-fatal coronary events were significantly reduced in the EPA plus statin group but in this study sudden cardiac death and coronary death did not differ between groups. Unstable angina was the main component contributing to the primary endpoint and this is a more subjective endpoint than other endpoints such as a myocardial infarction, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a limitation of the JELIS Study. Unfortunately, we do not have information on elderly patients (≥ 75 years).

 

REDUCE-IT

 

REDUCE-IT was a randomized, double-blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. mineral oil placebo in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established cardiovascular disease or high cardiovascular disease risk (diabetes plus one risk factor) who were on stable statin therapy (75). The primary end point was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina. At baseline, the median LDL-C level was 75.0 mg/dL, HDL-C level was 40.0 mg/dL, and TG level was 216.0 mg/dL. The median change in TG level from baseline to 1 year was a decrease of 18.3% (−39.0 mg/dL) in the EPA group and an increase of 2.2% (4.5 mg/dL) in the placebo group. After a median of 4.9 years the primary end-point occurred in 17.2% of the patients in the EPA group vs. 22.0% of the patients in the placebo group (HR 0.75; P<0.001), indicating a 25% decrease in events. In participants <65 years of age the primary end point was reduced by 35% (HR 0.65; 95% CI 0.54–0.78) while in participants ≥ 65 years of age the primary end point was reduced by 18% (HR 0.82; 95%CI 0.70–0.97; P Value for Interaction 0.06). The cardiovascular benefits of EPA were similar across baseline levels of TGs (<150, ≥150 to <200, and ≥200 mg per deciliter). Moreover, the cardiovascular benefits of EPA appeared to occur irrespective of the attained TG level at 1 year (≥150 or <150 mg/dL), suggesting that the ASCVD risk reduction was not associated with attainment of a normal TG level. Unfortunately, information on an elderly subgroup (≥ 75 years) is not available. 

 

It should be noted that in this trial mineral oil was used as the placebo. In the placebo group the LDL-C, non-HDL-C, and hsCRP levels were increased compared to the EPA group during the trial (LDL-C 96mg/dL vs 85mg/dL; non-HDL-C 130mg/dL vs. 113mg/dL; hsCRP 2.8mg/L vs. 1.8mg/L). The impact of these adverse changes on clinical outcomes is uncertain and whether they contributed to the apparent beneficial effects observed in the individuals treated with EPA is unknown.

 

STRENGTH TRIAL

 

The STRENGTH Trial was a double-blind, randomized, trial comparing 4 grams per day of a carboxylic acid formulation of omega-3 fatty acids (EPA and DHA; Epanova) (n = 6,539)) vs. corn oil placebo (n = 6539) in statin-treated participants with high cardiovascular risk, hypertriglyceridemia, and low levels of HDL-C (76). Approximately 55% of patients had established cardiovascular disease and approximately 70% had diabetes. Median LDL-C level was 75.0 mg/dL, median TG level was 240 mg/dL, and median HDL-C level was 36 mg/dL. There were minimal differences in the change in LDL-C and HDL-C levels between the treated and placebo groups after treatment for 12 months but as expected there was a greater reduction in TG levels in the group treated with omega-3-fatty acids (−19.0% vs −0.9%). The primary endpoint was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina requiring hospitalization which occurred in 12.0% of individuals treated with omega-3 CA vs. 12.2% treated with corn oil (HR 0.99; P = .84). There were no significant differences between the treatment groups with regard to the risk of the individual components of the primary end point over the 3-4 years of the study. Thus, in contrast to the JELIS and REDUCE-IT trials the STRENGTH trial did not demonstrate a benefit of treatment with a mixture of omega-3-fatty acids (EPA + DHA).

 

OMEMI TRIAL

 

The OMEMI trial was a randomized trial of 1.8 grams per day of omega-3-fatty acids (930 mg EPA and 660 mg DHA) (n= 505) vs. corn oil placebo (n= 509) in patients aged 70 to 82 years with a recent myocardial infarction (2-8 weeks) (77). Baseline LDL-C was approximately 76mg/dL, HDL-C was 49mg/dL, and TGs 110mg/dL. The primary endpoint was a composite of nonfatal myocardial infarction, unscheduled revascularization, stroke, all-cause death, and heart failure hospitalization after 2 years of follow-up. The primary endpoint occurred in 21.4% of patients on omega-3-fatty acids vs. 20.0% on placebo (HR 1.08; P=0.60). TGs levels decreased 8.1% in the omega-3-fatty acid group and increased 5.1% in the placebo group (between group difference 13.2%; P<0.001) while changes in LDL-C were minimal in both groups. Thus, similar to the STRENGTH trial no benefits on cardiovascular disease were observed with EPA + DHA treatment.

 

SUMMARY

 

  • High dose EPA (JELIS and REDUCE-IT) reduced ASCVD outcomes while high dose EPA+DHA (STENGTH and OMEMI) did not decrease ASCVD outcomes.
  • The decrease in TG levels is not a major contributor to the beneficial effect of high dose EPA as the combination of high dose EPA+DHA lowers TG levels to the same degree as EPA alone without benefit. Additionally, the JELIS trial only lowered TG levels by 5% but nevertheless reduced ASCVD events. It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial with TG lowering making only a small contribution to the decrease in cardiovascular disease. Other actions of EPA, such as decreasing platelet function, anti-inflammation, decreasing lipid oxidation, stabilizing membranes, etc. could account for or contribute to the reduction in ASCVD events (78).
  • Whether EPA has special properties that resulted in the reduction in ASCVD events in the REDUCE-IT trial or there were flaws in the trial design (i.e., the use of mineral oil as the placebo) is uncertain and debated. It should be noted that in the REDUCE-IT trial LDL-C and non-HDL-C levels were increased by approximately 10% in the mineral oil placebo group (75). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (75). Finally, an increase in hsCRP (20-30%) and other biomarkers of atherosclerosis (oxidized LDL-C, IL-6, IL-1 beta, and lipoprotein-associated phospholipase A2) were noted in the mineral oil group (75,79). In the STRENGTH trial there were no differences in LDL-C, Non-HDL-C, HDL-C, Apo B, or hsCRP levels between the treated vs. placebo groups (76). Whether EPA has special properties compared to DHA leading to a reduction in cardiovascular events or the mineral oil placebo resulted in adverse changes increasing ASCVD in the placebo resulting in an artifactual decrease in the EPA group is debated (80,81). Ideally, another large randomized ASCVD trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would help resolve this controversy. In the meantime, clinicians will need to use their clinical judgement on whether to treat patients with modest elevations in TG levels with EPA (icosapent ethyl; Vascepa) balancing the potential benefits of treatment vs. the potential side effects.

 

Summary of Lipid Lowering Drug Studies

 

The above results clearly demonstrate that lowering LDL-C levels with statins, ezetimibe, or PCSK 9 monoclonal antibodies will reduce ASCVD events in elderly patients (≥ 75 years of age) with pre-existing cardiovascular disease. In elderly patients without cardiovascular disease (primary prevention) the available data does not definitively demonstrate a decrease in ASCVD events with statin or ezetimibe therapy but is suggestive of a benefit (note there are no primary prevention trials with PCSK9 inhibitors). Additional data is required to determine if bempedoic acid and icosapent ethyl reduce ASCVD events in patients ≥ 75 years of age.

 

Studies in Progress

 

Studies are currently underway to provide definitive information on whether statin therapy is beneficial as primary prevention in the elderly. STAREE (NCT02099123) is a multicenter randomized trial in Australia of atorvastatin 40mg vs. placebo in adults ≥ 70 years of age without cardiovascular disease and PREVENTABLE (NCT04262206) is a multicenter randomized trial in the USA of atorvastatin vs. placebo in adults ≥ 75 years of age without cardiovascular disease (82,83). Other trials in the elderly that are in progress include SCOPE (NCT03770312) which is a multicenter randomized trial in Korea of low intensity vs. high intensity statin therapy in adults 76-85 years of age without CVD and SITE (Statins In The Elderly) (NCT02547883) which is a trial in France of patients ≥ 75 years of age on statin therapy who will be randomized to continue statin therapy or stop statin therapy.

 

SIDE EFFECTS OF LIPID LOWERING DRUGS

 

In this section we will describe the potential side effects of lipid lowering drugs with an emphasis on side effects likely to be seen in the elderly. Elderly patients may be more susceptible to side effects due to decreased renal function, decreased drug metabolism by the liver, polypharmacy leading to drug interactions, and co-morbidities. For a detailed discussion of the side effects of lipid lowering drugs see the Endotext chapters entitled “Cholesterol Lowering Drugs” and “Triglyceride Lowering Drugs” (49,50).

 

Statins

 

An umbrella review of meta-analyses of observational studies and randomized controlled trials examined 278 unique non-CVD outcomes from 112 meta-analyses of observational studies and 144 meta-analyses of RCTs and found that the only adverse effects associated with statin therapy were the development of diabetes and muscle disorders (84). For a detailed discussion of the side effects of statin therapy a scientific statement from the American Heart Association provides a comprehensive review (85).

 

DIABETES

 

After many years of statin use it was recognized that statins increase the risk of developing diabetes. In a meta-analysis of 13 trials with over 90,000 subjects, there was a 9% increase in the incidence of diabetes during follow-up among subjects receiving statin therapy (86). All statins appear to increase the risk of developing diabetes. In comparisons of intensive vs. moderate statin therapy, Preiss et al observed that patients treated with intensive statin therapy had a 12% greater risk of developing diabetes compared to subjects treated with moderate dose statin therapy (87). Older subjects, obese subjects, and subjects with high glucose levels were at a higher risk of developing diabetes while on statin therapy. In the Prosper trial in elderly subjects (70-82 years of age; average age 75), diabetes developed in 6.6% of patients treated with pravastatin vs. 5.1% of patients in the placebo group (51). Thus, statins may be unmasking and accelerating the development of diabetes that would have occurred naturally in these subjects at some point in time. In patients without risk factors for developing diabetes, treatment with statins does not appear to greatly increase the risk of developing diabetes.

 

In balancing the benefits and risks of statin therapy it is important to recognize that an increase in plasma glucose levels is a surrogate marker for an increased risk of developing micro and macrovascular disease (i.e., an increase in plasma glucose per se is not an event but rather increases the risk of future events). In contrast, statin therapy is preventing actual clinical events that cause morbidity and mortality. Furthermore, it may take many years for an elevated blood glucose to induce diabetic complications while the reduction in cardiovascular events with statin therapy occurs relatively quickly (after one-year benefits are seen). Finally, the number of patients needed to treat with statins to avoid one cardiovascular event is much lower (10-20 depending on the type of patient) than the number of patients needed to treat to cause one patient to develop diabetes (100–200 for one extra case of diabetes) (88). Patients on statin therapy, particularly those with risk factors for the development of diabetes, should be periodically screened for the development of diabetes with measurement of fasting glucose and/or HbA1c levels.

 

COGNITIVE DYSFUNCTION

 

Several randomized clinical trials have examined the effect of statin therapy on cognitive function and have not indicated any increased risk (for review see (89-92)). The Prosper Trial was designed to determine whether statin therapy will reduce cardiovascular disease in older subjects (age 70-82) (51). In this trial cognitive function was assessed repeatedly and no difference in cognitive decline was found in subjects treated with pravastatin compared to placebo (51,93). In the Heart Protection Study over 20,000 patients were randomized to simvastatin 40mg or placebo and again no significant differences in cognitive function was observed between the statin vs. placebo group (94). Additionally, a Cochrane review examined the effect of statin therapy in patients with established dementia and identified 4 studies with 1154 participants (95). In this analysis no benefit or harm of statin therapy on cognitive function could be demonstrated in this high-risk group of patients. Thus, randomized clinical trials do not indicate a significant association between statins and cognitive function.

 

MUSCLE

 

The most common side effect of statin therapy is muscle symptoms and many patients will discontinue the use of statins due to muscle symptoms. These can range from life threatening rhabdomyolysis, which is very rare, to myalgias, which are a common complaint (96). The risk of serious muscle disorders due to statin therapy is very small, particularly if one is aware of the potential drug interactions that increase the risk. In a case-control study with a cohort of 252,460 new users of lipid-lowering medications in U.S. health plans 21 cases of rhabdomyolysis were compared to 200 controls without rhabdomyolysis (97). Statin users >65 years of age had four times the risk of hospitalization for rhabdomyolysis than those under age 65.

The Cholesterol Treatment Trialists analyzed individual participant data on the development of muscle symptoms from 19 double-blind trials of statin versus placebo with 123,940 participants and four double-blind trials of a more intensive vs. a less intensive statin regimen with 30,724 participants (98). After a median follow-up of 4.3 years 27.1% of the individuals taking a statin vs. 26.6% on placebo reported muscle pain or weakness representing a 3% increase greater than placebo (risk ratio- 1.03; 95% CI 1.01-1.06) (Table 5). The specific muscle symptoms caused by statin therapy, myalgia, muscle cramps or spasm, limb pain, other musculoskeletal pain, or muscle fatigue or weakness were similar to those caused by placebo. The slight increase in muscle symptoms in the statin treated individuals was manifest in the first year of therapy but in the later years muscle symptoms were similar in the statin treated and placebo groups. The relative risk of statin induced muscle symptoms was greater in women than men. Intensive statin treatment with 40-80 mg atorvastatin or 20-40 mg rosuvastatin resulted in a higher risk of muscle symptoms than less intensive or moderate-intensity regimens but different statins at equivalent LDL-C lowering doses had similar effects on muscle symptoms. As shown in figure 7 muscle pain or weakness was slightly increased in patients > 65 years of age and similar in patients > 65 and ≤ 75 and those > 75 years of age. It should be noted that in individuals > 75 years of age the occurrence of muscle pain or weakness occurred in 39.6% of the individuals on the placebo, demonstrating the very high occurrence of muscle symptoms in this age group.

 

This study demonstrates that there is a small increase in muscle symptoms that primarily manifests in the first year of therapy. Statin therapy caused approximately 11 additional complaints of muscle pain or weakness per 1000 patients during the first year, but little excess in later years. Of particularly note is that 26.6% of patients taking a placebo had muscle symptoms demonstrating a very high frequency of this clinical complaint (even higher in patients > 75). Given the high prevalence of muscle complaints and the small increase attributed to statins it is very difficult to determine if a muscle complaint is actually due to the statin, which presents great clinical difficulties in patient management.

 

Table 5. Effect of Statin vs. Placebo on Muscle Symptoms

Symptom

Statin Events

Placebo Events

RR (95% CI)

Myalgia

12.0%

11.7%

1·03 (0·99–1·08)

Other musculoskeletal pain

13.3

13.0

1·03 (0·99–1·08)

Any muscle pain

26.9%

26.3%

1·03 (1·01–1·06)

Any muscle pain or weakness

27.1%

26.6%

1·03 (1·01–1·06)

 

Figure 7. Occurrence of muscle pain or weakness in different age groups in the Cholesterol Treatment Trialists meta-analysis.

 

While the results of the randomized trials suggest that muscle symptoms are not frequently induced by statin therapy, in typical clinical settings a significant percentage of patients are unable to tolerate statins due to muscle symptoms (in many studies as high as 5-25% of patients) (99-101). Additionally, when patients know that they are taking a statin they are more likely to have muscle symptoms (i.e. the nocebo effect) (102). Clinically differentiating statin induced myalgias from placebo induced myalgias is difficult, as there are no specific symptoms, signs, or biomarkers that clearly distinguish between the two. Thus, while statin induced myalgias are a real entity careful studies have shown that in the majority of patients with “statin induced muscle symptoms” the symptoms are not actually due to statin therapy. In the clinic it is difficult to be certain whether the muscle symptoms are actually due to true statin intolerance or to other factors.

 

A detailed discussion of statin induced muscle symptoms and a clinical approach to this problem is presented in the Endotext chapter entitled “Cholesterol Lowering Drugs” (50). In the section “Patients with Statin Intolerance” in this chapter we discuss the clinical approach to treating these patients.

 

Ezetimibe

 

Ezetimibe has not demonstrated significant side effects.

 

PCSK9 Monoclonal Antibodies

 

In a subgroup of patients from the FOURIER trial a prospective study of cognitive function (EBBINGHAUS Study) was carried out and no significant differences in cognitive function was observed over a median of 19 months in the PCSK9 treated vs. placebo group (103).

 

An issue of concern is whether lowering LDL-C to very low levels has the potential to cause toxicity. In a number of the PCSK9 studies a significant number of patients had LDL-C levels < 25mg/dL. For example, in the Odyssey long term study 37% of patients on alirocumab had two consecutive LDL-C levels below 25mg/dL and in the Osler long term study in patients treated with evolocumab 13% had values below 25mg/dL (104,105). In these short term PCSK9 studies, toxicity from very low LDL-C levels has not been observed. Additionally, in patients with Familial Hypobetalipoproteinemia LDL levels can be very low and these patients do not have any major disorders other than hepatic steatosis, which is not mechanistically due to low LDL-C levels (106). Similarly, there are rare individuals who are homozygous for loss of function mutations in the PCSK9 gene and they also do not appear to have major medical issues (107). Finally, in a number of statin trials and the IMPROVE-IT trial (statin + ezetimibe) there have been patients with very low LDL-C levels and an increased risk of side effects has not been consistently observed in those patients (108-111). Thus, with the limited data available there does not appear to be a major risk from markedly lowering LDL-C levels.

 

Bempedoic Acid

 

In clinical trials, 26% of bempedoic acid-treated patients with normal baseline uric acid values experienced hyperuricemia one or more times versus 9.5% in the placebo group (package insert). In the CLEAR Outcomes trial elevated uric acid levels occurred in 10.9% of the patients on bempedoic acid compared to 5.6% taking the placebo (69). Elevations in blood uric acid levels may lead to the development of gout. Gout was reported in 1.5% of patients treated with bempedoic acid vs. 0.4% of patients treated with placebo. The risk for gout attacks were higher in patients with a prior history of gout (11.2% for bempedoic acid treatment vs. 1.7% in the placebo group) (package insert). In patients with no prior history of gout only 1% of patients treated with bempedoic acid and 0.3% of the placebo group had a gouty attack (package insert). In the CLEAR Outcomes trial gout was increased in the bempedoic acid group (3.1% vs. 2.1%) (69).

 

In clinical trials tendon rupture occurred in 0.5% of patients treated with bempedoic acid vs. 0% of placebo treated patients and involved the rotator cuff (the shoulder), biceps tendon, or Achilles tendon (package insert). Tendon rupture occurred within weeks to months of starting bempedoic acid and occurred more frequently in patients over 60 years of age, in those taking corticosteroid or fluoroquinolone drugs, in patients with renal failure, and in patients with previous tendon disorders. In the CLEAR Outcomes trial tendon rupture was similar in the bempedoic acid and placebo group (bempedoic acid 1.2% and placebo 0.9%) (69).

 

Bempedoic acid treatment resulted in a mean increase in serum creatinine of 0.05 mg/dL compared to baseline. Approximately 3.8% of patients treated with bempedoic acid had BUN levels that doubled vs. 1.5% in the placebo group and about 2.2% of patients treated with bempedoic acid had creatinine values that increased by 0.5 mg/dL vs. 1.1% in the placebo group (package insert). Renal function returned to baseline when bempedoic acid was discontinued. In the CLEAR Outcomes trial renal impairment was increased in the bempedoic acid group (11.5% vs.8.6%) as was the change from baseline creatinine (0.05±0.2 mg/dL vs. 0.01±0.2 mg/dL) (69).

 

Omega-3-Fatty Acids

 

 At very high doses, omega-3-fatty acids can inhibit platelets and prolong bleeding time. However, at the recommended doses this has not been a major clinical problem but nevertheless when patients are on anti-platelet drugs one should be alert for the possibility of bleeding problems (Package Inserts for Lovaza, Vascepa, and Epanova). In the REDUCE-IT trial serious bleeding events occurred in 2.7% of the patients in the icosapent ethyl group and in 2.1% in the placebo group (P=0.06) (75). There were no fatal bleeding events in either group and the rates of hemorrhagic stroke, serious central nervous system bleeding, and serious gastrointestinal bleeding were not significantly higher in the EPA group than in the placebo group. In the STRENGHT trial any bleeding events and major bleeding events were similar in the omega-3 fatty acid group and placebo group (76).  A recent review found no evidence for discontinuing the use of omega-3 fatty acid treatment before invasive procedures or when given in combination with other agents that affect bleeding (112).

 

An increase in new-onset atrial fibrillation was observed in the REDUCE-IT trial in the patients treated with icosapent ethyl 4 grams/day (5.3% vs. 3.9%) and in the STRENGTH trial in the patients treated with omega-3-fatty acids (2.2% vs 1.3%)

 

CURRENT GUIDELINES AND LDL-C GOALS

 

This section discusses guidelines as they pertain to elderly patients.

 

2018 AHA/ACC/Multi-Society Report

 

The following summarizes the 2018 AHA/ACC guidelines (2).

 

PRIMARY PREVENTION

 

  • For individuals >75 years of age, randomized controlled trials of statin therapy do not provide strong evidence for benefit, so clinical assessment of risk status in a clinician–patient risk discussion is needed for deciding whether to continue or initiate statin treatment.
  • In individuals ≥ 75 years of age with an LDL-C level of 70 to 189mg/dL, initiating a moderate-intensity statin may be reasonable. Goal is to reduce LDL-C by 30-49% (note these guidelines recommend percent reduction rather than absolute LDL-C goals).
  • In individuals ≥ 75 years of age it may be reasonable to stop statin therapy when functional decline (physical or cognitive), multimorbidity, frailty, or reduced life-expectancy limits the potential benefits of statin therapy.
  • A shared decision-making process between clinicians and patients that individualizes decisions is indicated, with regular periodic reassessment.
  • Determining coronary artery calcium (CAC) score will help in determining which patients will benefit the most. For older adults with CAC scores of zero, the likelihood of benefits from statin therapy does not outweigh the risks. Limiting statin therapy to those with CAC scores greater than zero, combined with clinical judgment and patient preference, could provide a valuable awareness with which to inform shared decision-making.

 

SECONDARY PREVENTION

 

  • In patients ≥75 years of age with clinical ASCVD, it is reasonable to initiate moderate- or high-intensity statin therapy after evaluation of the potential for ASCVD risk reduction, adverse effects, and drug–drug interactions, as well as patient frailty and patient preferences. The goal of moderate statin therapy is to reduce LDL-C by 30-49% and the goal of high-intensity statin therapy is to reduce LDL-C by ≥ 50%. In very high-risk patients, a goal of an LDL-C < 70mg/dL and non-HDL-C < 100mg/dL is reasonable.
  • In patients ≥75 years of age who are tolerating high-intensity statin therapy, it is reasonable to continue high-intensity statin therapy after evaluation of the potential for ASCVD risk reduction, adverse effects, and drug-drug interactions, as well as patient frailty and patient preferences.

 

PATIENTS WITH DIABETES

 

  • In patients ≥ 75 years of age with diabetes mellitus and who are already on statin therapy, it is reasonable to continue statin therapy.
  • In patients ≥ 75 years of age with diabetes mellitus without cardiovascular disease it may be reasonable to start moderate statin therapy after a clinician-patient discussion of the potential benefits and risks of therapy. The goal is to decrease LDL-C by 30-49%.

 

2019 ESC/EAS Guidelines

 

The following summarizes the 2019 ESC/EAS guidelines (3).

 

  • Treatment with statins is recommended for older people with ASCVD in the same way as for younger patients.
  • Treatment with statins is recommended for primary prevention, according to the level of risk, in older people aged ≤ 75 years.
  • Initiation of statin treatment for primary prevention in older people aged >75 years may be considered, if at high-risk or above.
  • It is recommended that the statin is started at a low dose if there is significant renal impairment and/or the potential for drug interactions, and then titrated upwards to achieve LDL-C treatment goals.
  • In patients at very-high risk in primary or secondary prevention the goal is a 50% reduction in LDL-C and an LDL-C < 55mg/dL.
  • In patients at high risk in primary or secondary prevention the goal is a 50% reduction in LDL-C and an LDL-C < 70mg/dL.

The ESC/EAS criteria for very high risk and high risk are shown in table 6.

 

Table 6. ESC/EAS Criteria for Very-High Risk and High Risk for ASCVD Events

Very High Risk

Documented ASCVD or unequivocal on imaging

DM with target organ damage or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

Severe CKD (eGFR <30 mL/min/1.73 m2)

A calculated SCORE ≥ 10% for 10-year risk of fatal CVD

Familial Hypercholesterolemia with ASCVD or with another major risk factor

High Risk

Markedly elevated single risk factors, in particular LDL-C >190 mg/dL or BP ≥ 180/110 mmHg

Patients with Familial Hypercholesterolemia without other major risk factors

Patients with DM without target organ damage, with DM duration ≥ 10 years, or another additional risk factor

Moderate CKD (eGFR 30-59 mL/min/1.73 m2)

A calculated SCORE ≥ 5% and <10% for 10-year risk of fatal CVD.

 

Our Approach

 

Our approach is based on concepts taken from both the ACC/AHA and ESC/EAS guidelines (i.e., we try to utilize the best ideas from each guideline). There are several general principles regarding lipid lowering therapy that should be considered in deciding who to treat (113).

 

  • The higher the LDL-C level the greater the benefit of lowering LDL-C.
  • The greater the decrease in LDL-C the greater the benefit.
  • The higher the absolute risk of ASCVD the greater the benefit of lowering LDL-C.

 

Additional factors that need to be considered, particularly in elderly patients, include

 

  • Life expectancy.
  • Competing non-cardiovascular disorders.
  • Risk of drug side effects.
  • Potential for drug interactions.
  • Patient preferences.

 

PRIMARY PREVENTION

 

Given the absence of definitive outcome trials demonstrating the benefit of decreasing LDL-C levels in patients ≥ 75 years of age without cardiovascular disease one must use clinical judgement in deciding who to treat. It should be recognized, as discussed in detail earlier, that the available evidence suggests that decreasing LDL-C will reduce ASCVD events in the elderly. Our approach is to determine ASCVD risk and then balance the risk with competing factors such as life expectancy, non-cardiovascular disorders, potential for drug interactions, and patient preferences. We use the approach described below to determine risk.

 

Step 1- Calculate the 10-year risk of an ASCVD event using the AHA/ACC pooled cohort equation. In Europe one can use the SCORE OP risk prediction algorithms (114). This will provide an estimate of the risk of the patient having an ASCVD event/death.

Step 2- To gain further insight on the risk of ASCVD one can determine if patient has any risk enhancing factors (tables 7 and 8). This can help further stratify the patient’s risk.

Step 3- If after discussion with the patient, you and/or the patient is uncertain on the level of risk and the appropriate treatment plan obtaining a coronary calcium score (CAC) can be helpful. A CAC score of zero indicates low risk for ASCVD and allows one to not start statin therapy (2). Note that a CAC score of zero in cigarette smokers, patients with diabetes mellitus, those with a strong family history of ASCVD, and possibly chronic inflammatory conditions such as HIV, may still be associated with a substantial 10-year risk (2).

 

Following these steps, we can estimate the risk for ASCVD events and in conjunction with the general principles described above discuss with the patient a treatment plan. If the decision is to treat our goal is often an LDL-C < 100mg/dL and non-HDL-C < 130mg/dL but in high-risk patients our goal may be an LDL-C < 70mg/dL and non-HDL-C < 100mg/dL.

 

Table 7. Risk-Enhancing Factors

 Family history of premature ASCVD (males, age <55 y; females, age <65 y)

 Primary hypercholesterolemia (LDL-C ≥160mg/dL; non-HDL-C ≥190mg/dL

 Metabolic syndrome (increased waist circumference, elevated triglycerides [>175 mg/dL], elevated blood pressure, elevated glucose, and low HDL-C [<40 mg/dL in men; <50 in women mg/dL] are factors; tally of 3 makes the diagnosis)

 Chronic kidney disease (eGFR 15–59 mL/min/1.73 m2 with or without albuminuria; not treated with dialysis or kidney transplantation)

 Chronic inflammatory conditions such as psoriasis, RA, or HIV/AIDS

 History of premature menopause (before age 40 y) and history of pregnancy-associated conditions that increase later ASCVD risk such as preeclampsia

 High-risk race/ethnicities (e.g., South Asian ancestry)

 Lipid/biomarkers: Associated with increased ASCVD risk

  Persistently* elevated, primary hypertriglyceridemia (≥175 mg/dL);

  If measured:

  1. Elevated high-sensitivity C-reactive protein (≥2.0 mg/L)

  2. Elevated Lp(a) ≥50 mg/dL or ≥125 nmol/L

  3. Elevated apoB ≥130 mg/dL

  4. ABI <0.9

 ABI= ankle-brachial index, RA= rheumatoid arthritis.

Modified from reference (2).

 

Table 8. Factors Modifying Systematic Coronary Risk Estimation Risks

Social deprivation: the origin of many of the causes of CVD.

Obesity and central obesity as measured by the body mass index and waist circumference, respectively.

Physical inactivity.

Psychosocial stress

Family history of premature CVD (men: <55 years and women: <60 years).

Chronic immune-mediated inflammatory disorder.

Major psychiatric disorders.

Treatment for HIV infection.

Atrial fibrillation.

Left ventricular hypertrophy.

Chronic kidney disease.

Obstructive sleep apnea syndrome.

Metabolic associated fatty liver disease.

Modified from reference (3).

 

PATIENTS WITH DIABETES

 

In patients ≥ 75 with diabetes without pre-existing cardiovascular our approach is very similar to that described for primary prevention. In addition to the risk enhancers listed in tables 7 and 8 there are specific diabetes risk enhancers that clinicians should factor in their decisions (table 9). Also, as noted above, in the presence of diabetes a zero CAC score is not as strong an indicator of low risk for ASCVD as in non-diabetics.

 

In patients with diabetes because they usually have multiple risk factors and are at high risk for ASCVD events our typical LDL-C goal is < 70mg/dL and non-HDL-C < 100mg/dL. In the rare situation where there are minimal other risk factors an LDL-C goal < 100mg/dL and non-HDL-C < 130mg/dL is reasonable. 

 

Table 9. Diabetes-Specific Risk Enhancers That Are Independent of Other Risk Factors

Long duration (≥10 years for type 2 diabetes or ≥20 years for type 1 diabetes)

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m

Retinopathy

Neuropathy

ABI <0.9

ABI= ankle-brachial index.

Modified from reference (2).

 

SECONDARY PREVENTION

 

Studies have shown that lowering LDL-C levels with statins, ezetimibe, and PCSK9 monoclonal antibodies reduces ASCVD events in older adults with ASCVD. Thus, unless there are contraindications older patients with ASCVD should be treated with lipid lowering drugs to reduce ASCVD events. In elderly patients we will often employ a modest statin dose (for example atorvastatin 10-20mg or rosuvastatin 5-10mg) in combination with ezetimibe 10mg and then increase the statin dose, if necessary, based on lipid levels and the patient tolerating the treatment regimen. At a minimum our goal is an LDL-C < 70mg/dL and a non-HDL-C level < 100mg/dL but we would prefer lower values (ideally LDL-C < 55mg/dL and non-HDL-C < 85mg/dL) if they can be achieved with a statin + ezetimibe. In very high-risk patients (table 10) our goal is an LDL-C < 55mg/dL and non-HDL-C < 85mg/dL and adding a PCSK9 inhibitor may be required to achieve these levels in some patients.

 

Table 10. Criteria for Very High Risk

Very high-risk includes a history of multiple major ASCVD events or one major ASCVD event and multiple high-risk conditions.

 

Major ASCVD Events

 Recent ACS (within the past 12 months)

 History of MI (other than recent ACS event)

 History of ischemic stroke

 Symptomatic peripheral arterial disease (history of claudication with ABI <0.85, or previous revascularization or amputation)

 

High-Risk Conditions

 Age ≥65 y

 Heterozygous familial hypercholesterolemia

 History of prior coronary artery bypass surgery or percutaneous coronary intervention outside of the major ASCVD event(s)

 Diabetes mellitus

 Hypertension

 CKD (eGFR 15-59 mL/min/1.73 m2)

 Current smoking

 Persistently elevated LDL-C (LDL-C ≥100 mg/dL [≥2.6 mmol/L]) despite maximally tolerated statin therapy and ezetimibe

 History of congestive HF

ABI= ankle-brachial index; ACS= acute coronary syndrome.

Based on reference (2).

 

TREATMENT

 

Lifestyle

 

The lifestyle changes described below are recommended for all adults and are not specific for elderly individuals or for individuals with cardiovascular disease. The lifestyle changes recommended will lower lipid levels and are likely to reduce the risk of ASCVD.

 

EXERCISE

 

There is little debate that exercise is beneficial and that all individuals should be physically active. It is recommended that individuals participate in at least 150 minutes of moderate-intensity aerobic physical activity (for example 30 minutes 5 times per week) or 75 minutes per week of vigorous-intensity physical activity (115,116). Additionally, it is recommended that individuals participate in 2 days per week of muscle-strengthening activity (116). Because of the loss of muscle mass with aging it is very important to incorporate resistance training into the exercise program of elderly individuals.

 

A meta-analysis of exercise in the older individuals (>60 years of age) found that aerobic exercise decreased triglyceride and LDL-C levels and increased HDL-C levels while resistance exercise decreased LDL-C levels (117). Exercise also increases fitness and helps with weight loss. It should be noted that many elderly individuals may have substantial medical and social barriers to participating in exercise programs. Comorbidities, such as osteoarthritis, may limit exercise tolerance and make exercise challenging. Older individuals should be encouraged to be as active as possible.

 

DIET

 

For a detailed discussion of the effect of diet on lipids, lipoproteins and ASCVD see the Endotext chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels (118). There is general agreement on what constitutes a healthy diet and a brief summary of the Guidelines for Americans 2020-2025 is shown in table 11 and the guidelines from the American College of Cardiology/American Heart Association are shown in table 12.

 

Table 11. Guidelines for Americans 2020-2025

Recommend

Limit

Vegetables of all types—dark green; red and orange; beans, peas, and lentils; starchy; and other vegetables

Added sugars—Less than 10 percent of calories per day

Fruits, especially whole fruit

Saturated fat—Less than 10 percent of calories per day

Grains, at least half of which are whole grain

Sodium—Less than 2,300 milligrams per day

Dairy, including fat-free or low-fat milk, yogurt, and cheese, and/or lactose-free versions and fortified soy beverages and yogurt as alternatives

Alcoholic beverages—Adults can

choose not to drink or to drink in moderation by limiting intake to 2 drinks or less in a day for men and 1 drink or less in a day for women

Protein foods, including lean meats, poultry, and eggs; seafood; beans, peas, and lentils; and nuts, seeds, and soy products

 

Oils, including vegetable oils and oils in food, such as seafood and nuts

 

Full guideline is available at DietaryGuidelines.gov

 

Table 12. ACC/AHA Dietary Recommendations to Reduce Risk of ASCVD (115)

1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish is recommended

2. Replacement of saturated fat with dietary monounsaturated and polyunsaturated fats can be beneficial

3. A diet containing reduced amounts of cholesterol and sodium can be beneficial

4. As a part of a healthy diet, it is reasonable to minimize the intake of processed meats, refined carbohydrates, and sweetened beverages

5. As a part of a healthy diet, the intake of trans fats should be avoided

 

A summary of the effect of individual dietary constituents on lipid and lipoprotein levels is shown in table 13 (118). This table summarizes the results of numerous randomized trials examining the effect of dietary manipulations on lipid and lipoprotein levels.

 

Table 13. Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

Carbohydrates

Increase TGs, increase greater with simple sugars particularly fructose

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

SFA= saturated fatty acids, MUFA= monounsaturated fatty acids, PUFA= polyunsaturated fatty acids, TFA= trans fatty acids.

 

There is a huge literature describing the effect of diet on the risk of ASCVD and this literature is often conflicting and controversial (118). Several well recognized investigators have discussed the limitations of the information linking various diets and dietary constituents and the risk of disease (119,120). The major problem is that almost all of the information is based on observational studies and well conducted randomized trials measuring important ASCVD outcomes are very rare. Observational studies can demonstrate associations but do not definitively indicate that there is a cause-and-effect relationship. Unrecognized confounding variables can result in false associations.

 

Some of the more recent randomized dietary trials that have examined the effect of diet on ASCVD events are described below. For a discussion of other studies see reference (118). The PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) reduced the incidence of major ASCVD events (121). In this multicenter trial, carried out in Spain, over 7,000 patients at high risk for developing ASCVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. The average age of participants in this trial was 67. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary end point (MI, stroke, and death from ASCVD). Subgroup analysis demonstrated that the Mediterranean diet was equally beneficial in patients < 70 and ≥ 70 years of age. The Mediterranean diet resulted in only a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels, suggesting that the beneficial effects were not mediated by changes in lipids (122). The CORDIOPREV study and the Lyon Diet Heart Study were randomized trials that demonstrated that a Mediterranean diet reduces ASCVD events in patients with cardiovascular disease (secondary prevention) (123,124). Unfortunately, these studies did not have a sufficient number of patients > 70 years of age for analysis of the effect of the diet in older patients with pre-existing cardiovascular disease.

 

The results of these three randomized trials indicate that following a Mediterranean type diet reduces ASCVD. It is likely that the beneficial effects of the Mediterranean diet on ASCVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution.

 

LIPID LOWERING DRUGS

 

Current guidelines and lipid lowering goals are discussed in the guidelines section above. In this section we will focus on clinical decisions regarding the use of lipid lowering drugs. To maximize benefits of lipid lowering therapy we think it is important to achieve LDL-C goals.

 

Elderly Patients on Lipid Lowering Therapy

 

In elderly patients on lipid lowering therapy, we usually continue therapy if they are tolerating the medications without side effects. We will periodically check a lipid panel to make sure that they are achieving the goals of therapy. If not, we will adjust the lipid lowering medications to achieve the desired LDL-C goals. We will make changes in therapy if circumstances change. For example, if a patient develops metastatic cancer and is transferred to palliative or Hospice care we will stop the lipid lowering therapy. Similarly, if a new drug is required the current lipid lowering drugs may need to be changed to avoid drug interactions. Thus, in most patients continuing lipid lowering therapy is appropriate.

 

Primary Prevention in Elderly Patients

 

In elderly patients we usually initiate therapy using moderate-intensity statin therapy if therapy is indicated as discussed above. We typically use either atorvastatin 10-20mg or rosuvastatin 5-10mg. Our reason for using these statins is that if one needs to lower LDL-C further we can just increase the dose of the statin and not need to start a new statin. In certain circumstances we might use another statin to avoid drug interactions (for example in a patient living with HIV we might use pitavastatin). After 6-12 weeks on statin therapy, we check a lipid panel and if the patient is having any medication side effects. If the LDL-C is not at goal we will either increase the statin dose or if we feel that the patient is at risk for statin toxicity add ezetimibe 10mg instead. In a healthy elderly patient at high risk for ASCVD (for example high LDL-C, diabetes, and hypertension) we do not hesitate to use high-intensity statin therapy (atorvastatin 40-80mg and rosuvastatin 20-40mg) plus ezetimibe 10mg to achieve the LDL-C goal.

 

Secondary Prevention in Elderly Patients

 

Unless than is a contraindication we frequently start these patients on high-intensity statin therapy. After 6-12 weeks on statin therapy, we check a lipid panel and if the patient is having any medication side effects. We will often add ezetimibe as studies have shown that the greater the lowering of LDL-C the greater the reductions in ASCVD events. Additionally, ezetimibe is generic (i.e. inexpensive) and doesn’t typically cause side effects. We will use PCSK9 inhibitors following the principle that the higher the LDL-C and the greater the risk of ASCVD events the greater the cost effectiveness of using expensive PCSK9 inhibitors.

 

Mixed Hyperlipidemia

 

In patients with mixed hyperlipidemia (elevated LDL-C and triglyceride levels) Initial drug therapy should also be a statin unless triglyceride levels are greater than 500-1000mg/dL. If triglycerides are > 500-1000mg/dL initial therapy is directed at lowering triglyceride levels (49,125). In addition to lowering LDL-C levels, statins are also effective in lowering triglyceride levels particularly when the triglycerides are elevated. If LDL-C is not lowered sufficiently ezetimibe is a reasonable next step. The approach to the patient whose LDL-C levels are at goal but the triglycerides and non-HDL-C are still elevated is not clearly defined. As discussed above studies have failed to demonstrate that adding a fibrate or niacin reduces ASCVD events. The REDUCE-IT trial has demonstrated that icosapent ethyl (Vascepa) decreases ASCVD events in this patient population but as discussed in detail above the results of this study are debated because the mineral oil placebo increased LDL-C. non-HDL-C, hsCRP, and other biomarkers associated with an increased risk of ASCVD events. It is debated by various experts whether the beneficial effect seen in this study was due to the positive effects of icosapent ethyl or to negative effects of the placebo. Clinicians will need to use their clinical judgement on whether to treat patients with elevations in TG and non-HDL-C levels with icosapent ethyl balancing the potential benefits of treatment vs. the potential side effects. In making this decision in our elderly patients it is worth noting that in participants <65 years of age the primary end point was reduced by 35% (HR 0.65; 95% CI 0.54–0.78) while in participants ≥ 65 years of age the primary end point was reduced by 18% (HR 0.82; 95%CI 0.70–0.97; P Value for Interaction 0.06). Information on patients ≥ 75 years of age is not available.  

 

Patients with Statin Intolerance

 

Statin intolerance is frequently due to myalgias but on occasion can be due other issues, such as increased liver or muscle enzymes, cognitive dysfunction, or other neurological disorders. The percentage of patients who are “statin intolerant” varies greatly but in clinical practice a significant number of patients have difficulty taking statins.

 

It can be difficult to determine if the muscle symptoms that occur when a patient is taking a statin are actually due to the statin or are unrelated to statin use. The first step in a “statin intolerant patient” is to take a careful history of the nature and location of the muscle symptoms and the timing of onset in relation to statin use to determine whether the presentation fits the typical picture for statin induced myalgias. The characteristic findings with a statin induced myalgia are shown in table 14 and findings that are not typical for statin induced myalgia are shown in table 15. The disappearance of symptoms within a few weeks of stopping statins and the reappearance after restarting statins is very suggestive of the symptoms being due to true statin intolerance. An on-line tool (htpp://tools.acc.org/statinintolerance/#!/) and an app produced by the ACC/AHA are available. This tool characterizes patients based on 8 criteria into possible vs. unlikely to have statin induced muscle symptoms (table 16).

 

Table 14. Characteristic Findings with Statin Induced Myalgia

Symmetric

Proximal muscles

Muscle pain, tenderness, weakness, cramps

Symptom onset < 4 weeks after starting statin or dose increase

Improves within 2-4 weeks of stopping statin

Cramping is unilateral and involves small muscles of hands and feet

Same symptoms occur with re-challenge within 4 weeks

 

Table 15. Symptoms Atypical in Statin Induced Myalgia

Unilateral

Asymmetric

Small muscles

Joint or tendon pain

Shooting pain, muscle twitching or tingling

Symptom onset > 12 weeks

No improvement after discontinuing statin

 

Table 16. Diagnosis of Statin Associated Muscle Symptoms

Symptom timing

Symptom type

Symptom location

Sex

Age

Race/ethnicity

CK elevation > 5 times the upper limit of normal

Known risk factors for statin induced muscle symptoms and non-statin causes of muscle symptoms

 

One should also check a CK level but this is almost always in the normal range. If the CK is not elevated and the symptoms do not suggest a statin induced myalgia one can often reassure the patient and continue statin therapy. This is often successful and studies have shown that many patients that stop taking statins due to “statin induced myalgia” can be successfully treated with a statin (50). If the CK is elevated it should be repeated after instructing the patient to avoid exercise for 48 hours. Also, the CK levels should be compared to CK levels prior to starting therapy if available. If the CK remains elevated (3x upper limit of normal) the statin should be discontinued. Similarly, if the CK is normal but the symptoms are suggestive of a statin induced myalgia the statin should also be discontinued. The next step is to determine if one can identify reversible factors that could be increasing statin toxicity (hypothyroidism, drug interactions).  If none are identified the next step after the myalgias have resolved is to try a low dose of a different statin that is metabolized by a different pathway (for example instead of atorvastatin, which is metabolized by the CYP3A4 pathway, rosuvastatin, which has a different pathway of metabolism). Because statin side effects are dose related, a low dose of a statin may often be tolerated. One can also try several different statins as sometimes a patient may tolerate one statin and not others. A meta-analysis has shown that every other day administration of statins is as effective as daily administration in lowering lipid levels and therefore is a very reasonable strategy (126). In some instances, using a long-acting statin (rosuvastatin or atorvastatin) 1-3 times per week can work (we usually start with once per week and then slowly increase frequency as tolerated) (127). In these circumstances (low doses or 1-3 times per week) the reduction in LDL-C may not be sufficient but one can use combination therapy with other drugs such as ezetimibe, bempedoic acid, or PCSK9 inhibitors to achieve LDL-C target goals.

 

If after trying various approaches a patient still has myalgias and is unable to tolerate statin therapy one needs to utilize other approaches to lower LDL-C levels. We typically use the ezetimibe and bempedoic acid combination pill (Nexlizet), which can lower the LDL-C level by approximately 40%, which is often sufficient (128). If needed one could add a PCSK9 inhibitor to further decrease LDL-C.

 

CONCLUSION

 

ASCVD is a major cause of morbidity and mortality in elderly patients. In elderly patients with pre-existing ASCVD randomized clinical trials have shown that lipid lowering drug therapy with statins, ezetimibe, and PCSK9 inhibitors reduce ASCVD events. Thus, most elderly patients with ASCVD should be treated with lipid lowering drugs unless there are contraindications such as limited life expectancy, competing non-cardiovascular disorders, high risk of drug interactions or drug side effects. In elderly patients without ASCVD if they are already taking lipid lowering drugs and if they are tolerating the medications without side effects continuing therapy is usually reasonable as long as the clinical circumstances have not changed. In elderly patients not on lipid lowering therapy and without cardiovascular disease studies have suggested that lipid lowering therapy is beneficial but further studies are required to definitively demonstrate benefit. In these patients one needs to determine the patient’s risk for ASCVD events and then discuss the potential benefits and side effects with the patient to make a shared decision on whether to initiate therapy. Age per se should not be used to withhold therapy with lipid lowering drugs that can reduce the risk of ASCVD events.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education. The authors would like to thank Dan Streja, the original author of this chapter, who provided the framework for this updated chapter.

 

REFERENCES

 

  1. Savji N, Rockman CB, Skolnick AH, Guo Y, Adelman MA, Riles T, Berger JS. Association between advanced age and vascular disease in different arterial territories: a population database of over 3.6 million subjects. J Am Coll Cardiol 2013; 61:1736-1743
  2. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  3. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  4. Ghandehari H, Kamal-Bahl S, Wong ND. Prevalence and extent of dyslipidemia and recommended lipid levels in US adults with and without cardiovascular comorbidities: the National Health and Nutrition Examination Survey 2003-2004. Am Heart J 2008; 156:112-119
  5. Laurenzi M, Mancini M. Plasma lipids in elderly men and women. Eur Heart J 1988; 9 Suppl D:69-74
  6. Weijenberg MP, Feskens EJ, Kromhout D. Age-related changes in total and high-density-lipoprotein cholesterol in elderly Dutch men. Am J Public Health 1996; 86:798-803
  7. Abbott RD, Sharp DS, Burchfiel CM, Curb JD, Rodriguez BL, Hakim AA, Yano K. Cross-sectional and longitudinal changes in total and high-density-lipoprotein cholesterol levels over a 20-year period in elderly men: the Honolulu Heart Program. Ann Epidemiol 1997; 7:417-424
  8. Grunenberger F, Lammi-Keefe CJ, Schlienger JL, Deslypere JP, Hautvast JG. Longitudinal changes in serum lipids of elderly Europeans. SENECA Investigators. Eur J Clin Nutr 1996; 50 Suppl 2:S25-31
  9. Newschaffer CJ, Bush TL, Hale WE. Aging and total cholesterol levels: cohort, period, and survivorship effects. Am J Epidemiol 1992; 136:23-34
  10. Ettinger WH, Wahl PW, Kuller LH, Bush TL, Tracy RP, Manolio TA, Borhani NO, Wong ND, O'Leary DH. Lipoprotein lipids in older people. Results from the Cardiovascular Health Study. The CHS Collaborative Research Group. Circulation 1992; 86:858-869
  11. Curb JD, Reed DM, Yano K, Kautz JA, Albers JJ. Plasma lipids and lipoproteins in elderly Japanese-American men. J Am Geriatr Soc 1986; 34:773-780
  12. Wilson PW, Anderson KM, Harris T, Kannel WB, Castelli WP. Determinants of change in total cholesterol and HDL-C with age: the Framingham Study. J Gerontol 1994; 49:M252-257
  13. Garry PJ, Hunt WC, Koehler KM, VanderJagt DJ, Vellas BJ. Longitudinal study of dietary intakes and plasma lipids in healthy elderly men and women. Am J Clin Nutr 1992; 55:682-688
  14. Ferrara A, Barrett-Connor E, Shan J. Total, LDL, and HDL cholesterol decrease with age in older men and women. The Rancho Bernardo Study 1984-1994. Circulation 1997; 96:37-43
  15. Katsanos CS. Clinical considerations and mechanistic determinants of postprandial lipemia in older adults. Adv Nutr 2014; 5:226-234
  16. Spitler KM, Davies BSJ. Aging and plasma triglyceride metabolism. J Lipid Res 2020; 61:1161-1167
  17. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  18. Zuliani G, Romagnoni F, Bollini C, Leoci V, Soattin L, Fellin R. Low levels of high-density lipoprotein cholesterol are a marker of disability in the elderly. Gerontology 1999; 45:317-322
  19. Ranieri P, Rozzini R, Franzoni S, Barbisoni P, Trabucchi M. Serum cholesterol levels as a measure of frailty in elderly patients. Exp Aging Res 1998; 24:169-179
  20. Pel-Littel RE, Schuurmans MJ, Emmelot-Vonk MH, Verhaar HJ. Frailty: defining and measuring of a concept. J Nutr Health Aging 2009; 13:390-394
  21. Wilson DP. Is Atherosclerosis a Pediatric Disease? In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  22. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. N Engl J Med 1998; 338:1650-1656
  23. McGill HC, Jr., McMahan CA, Malcom GT, Oalmann MC, Strong JP. Effects of serum lipoproteins and smoking on atherosclerosis in young men and women. The PDAY Research Group. Pathobiological Determinants of Atherosclerosis in Youth. Arterioscler Thromb Vasc Biol 1997; 17:95-106
  24. Tuzcu EM, Kapadia SR, Tutar E, Ziada KM, Hobbs RE, McCarthy PM, Young JB, Nissen SE. High prevalence of coronary atherosclerosis in asymptomatic teenagers and young adults: evidence from intravascular ultrasound. Circulation 2001; 103:2705-2710
  25. Enos WF, Holmes RH, Beyer J. Coronary disease among United States soldiers killed in action in Korea; preliminary report. J Am Med Assoc 1953; 152:1090-1093
  26. McNamara JJ, Molot MA, Stremple JF, Cutting RT. Coronary artery disease in combat casualties in Vietnam. JAMA 1971; 216:1185-1187
  27. Webber BJ, Seguin PG, Burnett DG, Clark LL, Otto JL. Prevalence of and risk factors for autopsy-determined atherosclerosis among US service members, 2001-2011. JAMA 2012; 308:2577-2583
  28. Newman WP, 3rd, Freedman DS, Voors AW, Gard PD, Srinivasan SR, Cresanta JL, Williamson GD, Webber LS, Berenson GS. Relation of serum lipoprotein levels and systolic blood pressure to early atherosclerosis. The Bogalusa Heart Study. N Engl J Med 1986; 314:138-144
  29. McGill HC, Jr., McMahan CA. Determinants of atherosclerosis in the young. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Am J Cardiol 1998; 82:30T-36T
  30. Raitakari OT, Juonala M, Kahonen M, Taittonen L, Laitinen T, Maki-Torkko N, Jarvisalo MJ, Uhari M, Jokinen E, Ronnemaa T, Akerblom HK, Viikari JS. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study. JAMA 2003; 290:2277-2283
  31. Li S, Chen W, Srinivasan SR, Bond MG, Tang R, Urbina EM, Berenson GS. Childhood cardiovascular risk factors and carotid vascular changes in adulthood: the Bogalusa Heart Study. JAMA 2003; 290:2271-2276
  32. Davis PH, Dawson JD, Riley WA, Lauer RM. Carotid intimal-medial thickness is related to cardiovascular risk factors measured from childhood through middle age: The Muscatine Study. Circulation 2001; 104:2815-2819
  33. Juonala M, Magnussen CG, Venn A, Dwyer T, Burns TL, Davis PH, Chen W, Srinivasan SR, Daniels SR, Kahonen M, Laitinen T, Taittonen L, Berenson GS, Viikari JS, Raitakari OT. Influence of age on associations between childhood risk factors and carotid intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study, the Childhood Determinants of Adult Health Study, the Bogalusa Heart Study, and the Muscatine Study for the International Childhood Cardiovascular Cohort (i3C) Consortium. Circulation 2010; 122:2514-2520
  34. Magnussen CG, Venn A, Thomson R, Juonala M, Srinivasan SR, Viikari JS, Berenson GS, Dwyer T, Raitakari OT. The association of pediatric low- and high-density lipoprotein cholesterol dyslipidemia classifications and change in dyslipidemia status with carotid intima-media thickness in adulthood evidence from the cardiovascular risk in Young Finns study, the Bogalusa Heart study, and the CDAH (Childhood Determinants of Adult Health) study. J Am Coll Cardiol 2009; 53:860-869
  35. Klag MJ, Ford DE, Mead LA, He J, Whelton PK, Liang KY, Levine DM. Serum cholesterol in young men and subsequent cardiovascular disease. N Engl J Med 1993; 328:313-318
  36. Gray L, Lee IM, Sesso HD, Batty GD. Blood pressure in early adulthood, hypertension in middle age, and future cardiovascular disease mortality: HAHS (Harvard Alumni Health Study). J Am Coll Cardiol 2011; 58:2396-2403
  37. Pletcher MJ, Vittinghoff E, Thanataveerat A, Bibbins-Domingo K, Moran AE. Young Adult Exposure to Cardiovascular Risk Factors and Risk of Events Later in Life: The Framingham Offspring Study. PLoS One 2016; 11:e0154288
  38. Stamler J, Daviglus ML, Garside DB, Dyer AR, Greenland P, Neaton JD. Relationship of baseline serum cholesterol levels in 3 large cohorts of younger men to long-term coronary, cardiovascular, and all-cause mortality and to longevity. JAMA 2000; 284:311-318
  39. Warden BA, Fazio S, Shapiro MD. Familial Hypercholesterolemia: Genes and Beyond. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  40. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  41. Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. J Lipid Res 2009; 50 Suppl:S172-177
  42. Lubin JH, Couper D, Lutsey PL, Woodward M, Yatsuya H, Huxley RR. Risk of Cardiovascular Disease from Cumulative Cigarette Use and the Impact of Smoking Intensity. Epidemiology 2016; 27:395-404
  43. Lubin JH, Albanes D, Hoppin JA, Chen H, Lerro CC, Weinstein SJ, Sandler DP, Beane Freeman LE. Greater Coronary Heart Disease Risk With Lower Intensity and Longer Duration Smoking Compared With Higher Intensity and Shorter Duration Smoking: Congruent Results Across Diverse Cohorts. Nicotine Tob Res 2017; 19:817-825
  44. Ding N, Sang Y, Chen J, Ballew SH, Kalbaugh CA, Salameh MJ, Blaha MJ, Allison M, Heiss G, Selvin E, Coresh J, Matsushita K. Cigarette Smoking, Smoking Cessation, and Long-Term Risk of 3 Major Atherosclerotic Diseases. J Am Coll Cardiol 2019; 74:498-507
  45. Morton JI, Lazzarini PA, Polkinghorne KR, Carstensen B, Magliano DJ, Shaw JE. The association of attained age, age at diagnosis, and duration of type 2 diabetes with the long-term risk for major diabetes-related complications. Diabetes Res Clin Pract 2022; 190:110022
  46. de Jong M, Woodward M, Peters SAE. Duration of diabetes and the risk of major cardiovascular events in women and men: A prospective cohort study of UK Biobank participants. Diabetes Res Clin Pract 2022; 188:109899
  47. Feingold KR. Role of Glucose and Lipids in the Atherosclerotic Cardiovascular Disease in Patients with Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  48. Zoungas S, Woodward M, Li Q, Cooper ME, Hamet P, Harrap S, Heller S, Marre M, Patel A, Poulter N, Williams B, Chalmers J. Impact of age, age at diagnosis and duration of diabetes on the risk of macrovascular and microvascular complications and death in type 2 diabetes. Diabetologia 2014; 57:2465-2474
  49. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  50. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  51. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, Ford I, Gaw A, Hyland M, Jukema JW, Kamper AM, Macfarlane PW, Meinders AE, Norrie J, Packard CJ, Perry IJ, Stott DJ, Sweeney BJ, Twomey C, Westendorp RG. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623-1630
  52. Cholesterol Treatment Trialists Colloboration. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019; 393:407-415
  53. Amarenco P, Kim JS, Labreuche J, Charles H, Abtan J, Bejot Y, Cabrejo L, Cha JK, Ducrocq G, Giroud M, Guidoux C, Hobeanu C, Kim YJ, Lapergue B, Lavallee PC, Lee BC, Lee KB, Leys D, Mahagne MH, Meseguer E, Nighoghossian N, Pico F, Samson Y, Sibon I, Steg PG, Sung SM, Touboul PJ, Touze E, Varenne O, Vicaut E, Yelles N, Bruckert E. A Comparison of Two LDL Cholesterol Targets after Ischemic Stroke. N Engl J Med 2020; 382:9
  54. Andersson NW, Corn G, Dohlmann TL, Melbye M, Wohlfahrt J, Lund M. LDL-C Reduction With Lipid-Lowering Therapy for Primary Prevention of Major Vascular Events Among Older Individuals. J Am Coll Cardiol 2023; 82:1381-1391
  55. Orkaby AR, Driver JA, Ho YL, Lu B, Costa L, Honerlaw J, Galloway A, Vassy JL, Forman DE, Gaziano JM, Gagnon DR, Wilson PWF, Cho K, Djousse L. Association of Statin Use With All-Cause and Cardiovascular Mortality in US Veterans 75 Years and Older. JAMA 2020; 324:68-78
  56. Kim K, Lee CJ, Shim CY, Kim JS, Kim BK, Park S, Chang HJ, Hong GR, Ko YG, Kang SM, Choi D, Ha JW, Hong MK, Jang Y, Lee SH. Statin and clinical outcomes of primary prevention in individuals aged >75 years: The SCOPE-75 study. Atherosclerosis 2019; 284:31-36
  57. Bergami M, Cenko E, Yoon J, Mendieta G, Kedev S, Zdravkovic M, Vasiljevic Z, Milicic D, Manfrini O, van der Schaar M, Gale CP, Badimon L, Bugiardini R. Statins for primary prevention among elderly men and women. Cardiovasc Res 2022; 118:3000-3009
  58. Lavie G, Hoshen M, Leibowitz M, Benis A, Akriv A, Balicer R, Reges O. Statin Therapy for Primary Prevention in the Elderly and Its Association with New-Onset Diabetes, Cardiovascular Events, and All-Cause Mortality. Am J Med 2021; 134:643-652
  59. O'Sullivan JL, Kohl R, Lech S, Romanescu L, Schuster J, Kuhlmey A, Gellert P, Yasar S. Statin Use and All-Cause Mortality in Nursing Home Residents With and Without Dementia: A Retrospective Cohort Study Using Claims Data. Neurology 2024; 102:e209189
  60. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RM. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  61. Ouchi Y, Sasaki J, Arai H, Yokote K, Harada K, Katayama Y, Urabe T, Uchida Y, Hayashi M, Yokota N, Nishida H, Otonari T, Arai T, Sakuma I, Sakabe K, Yamamoto M, Kobayashi T, Oikawa S, Yamashita S, Rakugi H, Imai T, Tanaka S, Ohashi Y, Kuwabara M, Ito H. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019; 140:992-1003
  62. Kim BK, Hong SJ, Lee YJ, Hong SJ, Yun KH, Hong BK, Heo JH, Rha SW, Cho YH, Lee SJ, Ahn CM, Kim JS, Ko YG, Choi D, Jang Y, Hong MK. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet 2022; 400:380-390
  63. Lee B, Hong SJ, Rha SW, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Kim HK, Kim U, Choi YJ, Lee YJ, Lee SJ, Ahn CM, Ko YG, Kim BK, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin plus ezetimibe vs high-intensity statin according to baseline LDL-C in the treatment of atherosclerotic cardiovascular disease: A post-hoc analysis of the RACING randomized trial. Atherosclerosis 2023; 386:117373
  64. Lee SH, Lee YJ, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Park KH, Lee JH, Choi YJ, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Combination Moderate-Intensity Statin and Ezetimibe Therapy for Elderly Patients With Atherosclerosis. J Am Coll Cardiol 2023; 81:1339-1349
  65. Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, Ballantyne CM, Somaratne R, Legg J, Wasserman SM, Scott R, Koren MJ, Stein EA. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1500-1509
  66. Sever P, Gouni-Berthold I, Keech A, Giugliano R, Pedersen TR, Im K, Wang H, Knusel B, Sabatine MS, O'Donoghue ML. LDL-cholesterol lowering with evolocumab, and outcomes according to age and sex in patients in the FOURIER Trial. Eur J Prev Cardiol 2021; 28:805-812
  67. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  68. Sinnaeve PR, Schwartz GG, Wojdyla DM, Alings M, Bhatt DL, Bittner VA, Chiang CE, Correa Flores RM, Diaz R, Dorobantu M, Goodman SG, Jukema JW, Kim YU, Pordy R, Roe MT, Sy RG, Szarek M, White HD, Zeiher AM, Steg PG. Effect of alirocumab on cardiovascular outcomes after acute coronary syndromes according to age: an ODYSSEY OUTCOMES trial analysis. Eur Heart J 2020; 41:2248-2258
  69. Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen CF, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353-1364
  70. AIM-HIGH Investigators, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255-2267
  71. Hps Thrive Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014; 371:203-212
  72. ACCORD Study Group, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  73. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med 2022; 387:1923-1934
  74. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 2007; 369:1090-1098
  75. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med 2019; 380:11-22
  76. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020; 324:2268-2280
  77. Kalstad AA, Myhre PL, Laake K, Tveit SH, Schmidt EB, Smith P, Nilsen DWT, Tveit A, Fagerland MW, Solheim S, Seljeflot I, Arnesen H. Effects of n-3 Fatty Acid Supplements in Elderly Patients After Myocardial Infarction: A Randomized, Controlled Trial. Circulation 2021; 143:528-539
  78. Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020; 40:1135-1147
  79. Ridker PM, Rifai N, MacFadyen J, Glynn RJ, Jiao L, Steg PG, Miller M, Brinton EA, Jacobson TA, Tardif JC, Ballantyne CM, Mason RP, Bhatt DL. Effects of Randomized Treatment With Icosapent Ethyl and a Mineral Oil Comparator on Interleukin-1beta, Interleukin-6, C-Reactive Protein, Oxidized Low-Density Lipoprotein Cholesterol, Homocysteine, Lipoprotein(a), and Lipoprotein-Associated Phospholipase A2: A REDUCE-IT Biomarker Substudy. Circulation 2022; 146:372-379
  80. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  81. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  82. Joseph J, Pajewski NM, Dolor RJ, Sellers MA, Perdue LH, Peeples SR, Henrie AM, Woolard N, Jones WS, Benziger CP, Orkaby AR, Mixon AS, VanWormer JJ, Shapiro MD, Kistler CE, Polonsky TS, Chatterjee R, Chamberlain AM, Forman DE, Knowlton KU, Gill TM, Newby LK, Hammill BG, Cicek MS, Williams NA, Decker JE, Ou J, Rubinstein J, Choudhary G, Gazmuri RJ, Schmader KE, Roumie CL, Vaughan CP, Effron MB, Cooper-DeHoff RM, Supiano MA, Shah RC, Whittle JC, Hernandez AF, Ambrosius WT, Williamson JD, Alexander KP. Pragmatic evaluation of events and benefits of lipid lowering in older adults (PREVENTABLE): Trial design and rationale. J Am Geriatr Soc 2023; 71:1701-1713
  83. Zoungas S, Curtis A, Spark S, Wolfe R, McNeil JJ, Beilin L, Chong TT, Cloud G, Hopper I, Kost A, Nelson M, Nicholls SJ, Reid CM, Ryan J, Tonkin A, Ward SA, Wierzbicki A. Statins for extension of disability-free survival and primary prevention of cardiovascular events among older people: protocol for a randomised controlled trial in primary care (STAREE trial). BMJ Open 2023; 13:e069915
  84. He Y, Li X, Gasevic D, Brunt E, McLachlan F, Millenson M, Timofeeva M, Ioannidis JPA, Campbell H, Theodoratou E. Statins and Multiple Noncardiovascular Outcomes: Umbrella Review of Meta-analyses of Observational Studies and Randomized Controlled Trials. Ann Intern Med 2018; 169:543-553
  85. Newman CB, Preiss D, Tobert JA, Jacobson TA, Page RL, 2nd, Goldstein LB, Chin C, Tannock LR, Miller M, Raghuveer G, Duell PB, Brinton EA, Pollak A, Braun LT, Welty FK. Statin Safety and Associated Adverse Events: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2019; 39:e38-e81
  86. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-742
  87. Preiss D, Seshasai SR, Welsh P, Murphy SA, Ho JE, Waters DD, DeMicco DA, Barter P, Cannon CP, Sabatine MS, Braunwald E, Kastelein JJ, de Lemos JA, Blazing MA, Pedersen TR, Tikkanen MJ, Sattar N, Ray KK. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 2011; 305:2556-2564
  88. Sattar N. Statins and diabetes: What are the connections? Best Pract Res Clin Endocrinol Metab 2023; 37:101749
  89. Rojas-Fernandez C, Hudani Z, Bittner V. Statins and Cognitive Side Effects: What Cardiologists Need to Know. Endocrinol Metab Clin North Am 2016; 45:101-116
  90. Rojas-Fernandez CH, Goldstein LB, Levey AI, Taylor BA, Bittner V. An assessment by the Statin Cognitive Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S5-16
  91. Richardson K, Schoen M, French B, Umscheid CA, Mitchell MD, Arnold SE, Heidenreich PA, Rader DJ, deGoma EM. Statins and cognitive function: a systematic review. Ann Intern Med 2013; 159:688-697
  92. Olmastroni E, Molari G, De Beni N, Colpani O, Galimberti F, Gazzotti M, Zambon A, Catapano AL, Casula M. Statin use and risk of dementia or Alzheimer's disease: a systematic review and meta-analysis of observational studies. Eur J Prev Cardiol 2022; 29:804-814
  93. Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM, Murphy MB, Ford I, Macfarlane PW, Sattar N, Packard CJ, Stott DJ, Shepherd J, Bollen EL, Blauw GJ, Jukema JW, Westendorp RG. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol 2010; 257:85-90
  94. Collins R, Armitage J, Parish S, Sleight P, Peto R, Heart Protection Study Collaborative Group. Effects of cholesterol-lowering with simvastatin on stroke and other major vascular events in 20536 people with cerebrovascular disease or other high-risk conditions. Lancet 2004; 363:757-767
  95. McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Statins for the treatment of dementia. Cochrane Database Syst Rev 2014:CD007514
  96. Rosenson RS, Baker SK, Jacobson TA, Kopecky SL, Parker BA, The National Lipid Association's Muscle Safety Expert Panel. An assessment by the Statin Muscle Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S58-71
  97. Schech S, Graham D, Staffa J, Andrade SE, La Grenade L, Burgess M, Blough D, Stergachis A, Chan KA, Platt R, Shatin D. Risk factors for statin-associated rhabdomyolysis. Pharmacoepidemiol Drug Saf 2007; 16:352-358
  98. Cholesterol Treatment Trialists Collaboration. Effect of statin therapy on muscle symptoms: an individual participant data meta-analysis of large-scale, randomised, double-blind trials. Lancet 2022; 400:832-845
  99. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients--the PRIMO study. Cardiovasc Drugs Ther 2005; 19:403-414
  100. Buettner C, Davis RB, Leveille SG, Mittleman MA, Mukamal KJ. Prevalence of musculoskeletal pain and statin use. J Gen Intern Med 2008; 23:1182-1186
  101. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208-215
  102. Gupta A, Thompson D, Whitehouse A, Collier T, Dahlof B, Poulter N, Collins R, Sever P. Adverse events associated with unblinded, but not with blinded, statin therapy in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid-Lowering Arm (ASCOT-LLA): a randomised double-blind placebo-controlled trial and its non-randomised non-blind extension phase. Lancet 2017; 389:2473-2481
  103. Giugliano RP, Mach F, Zavitz K, Kurtz C, Im K, Kanevsky E, Schneider J, Wang H, Keech A, Pedersen TR, Sabatine MS, Sever PS, Robinson JG, Honarpour N, Wasserman SM, Ott BR. Cognitive Function in a Randomized Trial of Evolocumab. N Engl J Med 2017; 377:633-643
  104. Koren MJ, Giugliano RP, Raal FJ, Sullivan D, Bolognese M, Langslet G, Civeira F, Somaratne R, Nelson P, Liu T, Scott R, Wasserman SM, Sabatine MS. Efficacy and safety of longer-term administration of evolocumab (AMG 145) in patients with hypercholesterolemia: 52-week results from the Open-Label Study of Long-Term Evaluation Against LDL-C (OSLER) randomized trial. Circulation 2014; 129:234-243
  105. Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M, Stroes ES, Langslet G, Raal FJ, El Shahawy M, Koren MJ, Lepor NE, Lorenzato C, Pordy R, Chaudhari U, Kastelein JJ. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1489-1499
  106. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  107. McKenney JM. Understanding PCSK9 and anti-PCSK9 therapies. J Clin Lipidol 2015; 9:170-186
  108. LaRosa JC, Grundy SM, Kastelein JJ, Kostis JB, Greten H. Safety and efficacy of Atorvastatin-induced very low-density lipoprotein cholesterol levels in Patients with coronary heart disease (a post hoc analysis of the treating to new targets [TNT] study). Am J Cardiol 2007; 100:747-752
  109. Wiviott SD, Cannon CP, Morrow DA, Ray KK, Pfeffer MA, Braunwald E. Can low-density lipoprotein be too low? The safety and efficacy of achieving very low low-density lipoprotein with intensive statin therapy: a PROVE IT-TIMI 22 substudy. J Am Coll Cardiol 2005; 46:1411-1416
  110. Everett BM, Mora S, Glynn RJ, MacFadyen J, Ridker PM. Safety profile of subjects treated to very low low-density lipoprotein cholesterol levels (<30 mg/dl) with rosuvastatin 20 mg daily (from JUPITER). Am J Cardiol 2014; 114:1682-1689
  111. Giugliano RP, Wiviott SD, Blazing MA, De Ferrari GM, Park JG, Murphy SA, White JA, Tershakovec AM, Cannon CP, Braunwald E. Long-term Safety and Efficacy of Achieving Very Low Levels of Low-Density Lipoprotein Cholesterol : A Prespecified Analysis of the IMPROVE-IT Trial. JAMA Cardiol 2017; 2:547-555
  112. Wachira JK, Larson MK, Harris WS. n-3 Fatty acids affect haemostasis but do not increase the risk of bleeding: clinical observations and mechanistic insights. Br J Nutr 2014; 111:1652-1662
  113. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  114. Score Op working group E. S. C. Cardiovascular risk collaboration. SCORE2-OP risk prediction algorithms: estimating incident cardiovascular event risk in older persons in four geographical risk regions. Eur Heart J 2021; 42:2455-2467
  115. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Munoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 140:e596-e646
  116. Piercy KL, Troiano RP, Ballard RM, Carlson SA, Fulton JE, Galuska DA, George SM, Olson RD. The Physical Activity Guidelines for Americans. JAMA 2018; 320:2020-2028
  117. Yun H, Su W, Zhao H, Li H, Wang Z, Cui X, Xi C, Gao R, Sun Y, Liu C. Effects of different exercise modalities on lipid profile in the elderly population: A meta-analysis. Medicine (Baltimore) 2023; 102:e33854
  118. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  119. Ioannidis JPA. The Challenge of Reforming Nutritional Epidemiologic Research. JAMA 2018; 320:969-970
  120. Nissen SE. U.S. Dietary Guidelines: An Evidence-Free Zone. Ann Intern Med 2016; 164:558-559
  121. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  122. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  123. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  124. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  125. Chait A, Feingold KR. Approach to patients with hypertriglyceridemia. Best Pract Res Clin Endocrinol Metab 2023; 37:101659
  126. Awad K, Mikhailidis DP, Toth PP, Jones SR, Moriarty P, Lip GYH, Muntner P, Catapano AL, Pencina MJ, Rosenson RS, Rysz J, Banach M. Efficacy and Safety of Alternate-Day Versus Daily Dosing of Statins: a Systematic Review and Meta-Analysis. Cardiovasc Drugs Ther 2017; 31:419-431
  127. Kennedy SP, Barnas GP, Schmidt MJ, Glisczinski MS, Paniagua AC. Efficacy and tolerability of once-weekly rosuvastatin in patients with previous statin intolerance. J Clin Lipidol 2011; 5:308-315
  128. Ballantyne CM, Laufs U, Ray KK, Leiter LA, Bays HE, Goldberg AC, Stroes ES, MacDougall D, Zhao X, Catapano AL. Bempedoic acid plus ezetimibe fixed-dose combination in patients with hypercholesterolemia and high CVD risk treated with maximally tolerated statin therapy. Eur J Prev Cardiol 2020; 27:593-603

 

The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels

ABSTRACT

 

The role of lipids and lipoproteins as causal factors for cardiovascular disease (CVD) is well established. Dietary saturated fatty acids (SFA), which are in milk, butter, cheese, beef, lamb, pork, poultry, palm oil, and coconut oil increase LDL-C and HDL-C. The increase in LDL-C is due to a decrease in hepatic LDL clearance and an increase in LDL production secondary to a decrease in hepatic LDL receptors. Monounsaturated fatty acids (MUFA) are in olive, canola, peanut, safflower, and sesame oil, and avocados, peanut butter, and many nuts and seeds and polyunsaturated fatty acids (PUFA) are in soybean, corn, and sunflower oil, and some nuts and seeds, tofu, and soybeans. Both MUFA and PUFA lower LDL-C by increasing hepatic LDL receptor activity. Dietary cholesterol is found in egg yolks, shrimp, beef, pork, poultry, cheese, and butter and increase LDL-C but the effect is modest and varies with approximately 15-25% of individuals being hyper-responders with more robust increases. Dietary cholesterol reduces hepatic LDL receptor activity, decreasing the clearance and increasing the production of LDL. Trans fatty acids (TFA) occur naturally in meat and dairy products and are formed during the partial hydrogenation of vegetable fat. TFA increase LDL-C and decrease HDL-C. Carbohydrates (CHO) can be divided into high-quality, for example fruits, legumes, vegetables, and whole grains, or low-quality, which include refined grains, starches, and added sugars. CHO increase TG with low quality CHO, particularly added sugars, having a more robust effect. Dietary CHO, particularly fructose, promotes hepatic de novo fatty acid synthesis leading to increased VLDL secretion. Fiber is found mostly in fruits, vegetables, whole and unrefined grains, nuts, seeds, beans, and legumes and phytosterols are naturally occurring constituents of plants and are found in vegetable oils, cereals, nuts, fruit and vegetables. Both dietary fiber and phytosterols decrease LDL-C by decreasing intestinal cholesterol absorption.

Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

CHO

Increase TGs particularly simple sugars

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

With regards to CVD there are very few well conducted randomized controlled trials and most of the information is derived from observational studies that demonstrate associations. These observational studies have found that fruits, vegetables, beans/legumes, nuts/seeds, whole grains, fish, yogurt, fiber, seafood omega-3 fatty acids, and polyunsaturated fats were associated with a decreased risk of CVD while unprocessed red meats, processed meats, sugar-sweetened beverages, high glycemic load CHO, and trans-fats were associated with an increased risk of CVD. Randomized trials have shown that a Mediterranean diet reduces CVD. Based on this information current guidelines for the general population recommend 1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish 2. Replacement of SFA with MUFA and PUFA 3. A reduced amount of dietary cholesterol 4. Minimizing intake of processed meats, refined CHO, and sweetened beverages and 5. Avoidance of TFA. For individuals with a high LDL-C limiting dietary SFA, TFA, and cholesterol and increasing fiber and phytosterols will help lower LDL-C while in individuals with high TG limiting low quality CHO, particularly simple sugars, and ethanol with weight loss, if indicated, will help lower TG.

 

INTRODUCTION

 

There is a huge literature describing the effect of diet on the risk of cardiovascular disease (CVD) and this literature is often conflicting and controversial. Several well recognized investigators have discussed the limitations of the information linking various diets and dietary constituents and the risk of disease (1,2). The major problem is that almost all of the information is based on observational studies and well conducted randomized trials measuring important cardiovascular outcomes are very rare. Observational studies can demonstrate associations but do not necessarily indicate that there is a cause-and-effect relationship. Unrecognized confounding variables can result in false associations. In several instances a robust association was observed in observation trials but randomized trials failed to confirm these observations (3). For example, several observational studies showed that higher vitamin E intake from dietary sources or supplements was associated with a lower risk of CVD (4-8), but randomized controlled trials failed to demonstrate a reduction in cardiovascular events with vitamin E supplementation (9-12). Observational studies have also reported that vitamin B6, B12, or folic acid intake reduced the risk of CVD (13-15), but again randomized controlled trials failed to demonstrate a benefit of increased vitamin intake on CVD (16-19). These results point to potential deficiencies in observational studies and the need to recognize that the associations demonstrated in observational studies may not always be causal. Therefore, in this chapter, where possible, we will focus on randomized controlled trials.

 

Moreover, even the interpretation of the results of observational trials is often debated. For example, a 2019 meta-analysis and systematic review published in the Annals of Internal Medicine reached the conclusion that “the magnitude of association between red and processed meat consumption and all-cause mortality and adverse cardiometabolic outcomes is very small, and the evidence is of low certainty” (20). This conclusion is contrary to the recommendations of almost all dietary guidelines and as would be expected this resulted in a critique challenging this conclusion (21). There are numerous other instances where there are conflicting results and interpretations in the literature linking diet with CVD making it difficult to sort out fact from fiction.

 

The information pertaining to the effect of dietary manipulations on lipid and lipoprotein levels are frequently based on randomized controlled trials rather than observational studies and therefore tend to be more consistent. However, even in these studies the results are sometimes conflicting. There are many factors that could account for this variability including the heterogeneity in study settings, type of individuals studied, study designs, differences in baseline diets, adherence to the study diet, differences in types of diet or dietary composition, methods and accuracy of the methods used to measure lipid and lipoprotein levels, and many other factors.

 

Additionally, the clinician should recognize that the lipid response of an individual patient to dietary manipulations can vary greatly, is very modest on average (in the range of 10% reductions, typically), and in most cases will not prevent the need for lipid lowering medications. The importance of genetic differences on these responses is often under recognized by patients and providers. For example, individuals with an apo E4 allele have a more robust decrease in LDL-C in response to a decrease in dietary fat and cholesterol than subjects carrying the apo E3 or apo E2 alleles (22). Polymorphisms in other genes have also been shown to modulate the lipid and lipoprotein response to dietary manipulations (22,23). Clinical conditions can also affect the response to diet. For example, the expected lipid and lipoprotein response to a low cholesterol, low saturated fatty acids (SFA) diet is blunted in obese individuals (24). Therefore, the effect of a specific diet can vary from individual to individual and the clinician will have to monitor a patient’s response.

 

It should be recognized that when one increases or decreases a particular macronutrient in the diet (lipids, carbohydrates (CHO), or protein) there needs to be a reciprocal change in another macronutrient to maintain caloric balance. It can therefore be difficult to know whether the increase in a particular nutrient or a decrease in another nutrient is accounting for the observed effect (for example decreasing SFA and increasing CHO). Where possible I will try to specify which nutrient was decreased and which was increased in the studies described.

 

Finally, it is important to look at the effect of diet on lipids independent of weight loss. Weight loss per se can affect lipid levels resulting in a decrease in triglycerides and LDL-C and an increase in HDL-C levels (25). For a detailed discussion of the effect of weight loss on lipid levels see the chapter on obesity and dyslipidemia (25).

 

In this chapter we will first discuss the effect of various macronutrients, then specific foods, and finally specific diets on lipids and lipoprotein levels.  

 

DIETARY SATURATED FATTY ACIDS

 

Major sources of saturated fatty acids (SFA) in the diet are milk, butter, cheese, other dairy products, beef, lamb, pork, poultry particularly the skin, palm oil, palm kernel oil, and coconut oil (tables 1 and 3).

 

Table 1. Fatty Acid Composition of Foods High in Saturated Fat

 

Total Fat

grams/100 grams

SFA

grams/100 grams

MUFA

grams/100 grams

PUFA

grams/100 grams

Hamburger

15.0

5.89

6.66

0.49

Pork loin

13.3

5.23

6.19

1.20

Chicken

12.6

3.50

4.93

2.74

Lamb

15.1

6.90

7.00

1.20

Whole milk*

3.9

2.5

1.0

0.1

Gouda cheese**

30.6

20.3

7.4

0.9

Butter***

82.2

52.1

20.9

2.8

*TFA = 0.1g/100g; **TFA = 1.1g/100g; TFA = 2.9g/100g.

TFA= trans fatty acids, MUFA= monounsaturated fatty acids, PUFA= polyunsaturated fatty acids.

 

Effect of Dietary Saturated Fatty Acids on Cardiovascular Disease

 

OBSERVATIONAL STUDIES

 

Dietary guidelines uniformly recommend reducing the intake of SFA. There are a large number of observational trials that have shown an association between dietary SFA intake and CVD (26-31). However, there are meta-analyses that have not found an association between dietary SFA intake and CVD (32-36). A possible explanation for this discordance is whether the SFA in the diet is replaced by polyunsaturated fatty acids (PUFA) vs. replaced by CHO. When SFA is replaced by PUFA there is a reduction in CVD whereas replacement with CHO has no benefit on CVD (27-29,37-39). However, replacement of SFA with high quality CHO may be beneficial (27,37,38). Additionally, in one study SFA from meat was associated with an increased risk of CVD while SFA from dairy products was associated with a decrease in CVD (40). Thus, the source of SFA may be important. 

 

As noted above, observational studies can demonstrate an association but are not able to definitively demonstrate a causal relationship. It is therefore essential to review the results of randomized controlled trials on the effect of decreasing dietary SFA on preventing cardiovascular events.

 

RANDOMIZED CONTROLLED OUTCOME TRIALS

 

This section will review the major randomized trials analyzing the effect of decreasing SFA intake on preventing CVD. Studies with very few participants, few cardiovascular events, or very short-term studies will not be included. It is important to note that many of these studies were carried out in the 1950’s and 1960’s when the diagnosis and treatment of CVD was very primitive compared to current standards. Also, typical diets were much different (higher in SFA) and mean plasma cholesterol levels were higher. Lastly, the methodology of these studies was not up to the current standards by which randomized controlled trials are performed (small number of patients, often not blinded, inadequate statistical power, non-specific endpoints, etc.). Thus, the accuracy of these trials and the relevancy of these older studies to current times is uncertain.

 

In a study from England initiated in 1957, 252 men under the age of sixty-five who recently

had a myocardial infarction were assigned to a low-fat diet or usual diet (41). The low-fat diet was limited to 40 grams per day of fat with decreases in butter and meat. The intake of fat during the trial was approximately 100-120 grams per day in the usual diet group and slightly greater than 40 grams per day in the low-fat diet group. At the time of the study the typical diet was high in SFA so a decrease in total fat would have resulted in a significant decrease in SFA. During the trial serum cholesterol levels were in the 240mg/dL range in the usual diet group and 220mg/dL in the low-fat diet group. There were no differences between the two groups in cardiovascular events during the 5 years of the trial. To see a reduction in cardiovascular events with the modest reduction in serum cholesterol levels this study would have required a much larger number of participants.

 

The Oslo Diet-Heart Study randomized men under 65 years of age with a history of a myocardial infarction to a diet low in SFA and cholesterol, and high in PUFA (n=206) or their usual diet (n=206) (42). Cholesterol levels were approximately 295mg/dL and decreased to approximately 240mg/dL in the patients on the low SFA diet with minimal changes in the control group. After 5 years major cardiovascular events and cardiovascular mortality were reduced in the group on the low SFA diet (Events- 61 low SFA group vs. 81 control group; Mortality- 38 low SFA group vs. 52 control group).

 

The Medical Research Council soya-bean trial randomized men under 60 years of age with a recent myocardial infarction to continue their usual diet (n=194) or a diet low in SFA and containing 85 grams of soya-bean oil daily (PUFA) (n=199) (43). The low SFA diet lowered cholesterol from 272 to 213mg/dL (22% decrease) while in the controls, cholesterol decreased from 273 to 259mg/dL (6% decrease). The primary outcome was first relapse (myocardial infarction, angina, sudden death). After 4 years, 62 of 199 in the soybean oil group had a recurrent coronary event compared with 74 of 194 in the usual diet group; the difference, −18% (95% CI, −38 to 7), was not statistically significant but given the small number of participants was suggestive of benefit.

 

The Los Angeles Veterans Administration Center study randomized 422 men to the conventional control diet and 424 to the experimental diet low in SFA and cholesterol and enriched in PUFA (44,45). 30% of the men had CVD. The baseline plasma cholesterol was 233mg/dL and on treatment there was a 13% decrease in the experimental diet compared to controls. Over 8 years the primary endpoint of myocardial infarction and sudden death from ischemia were reduced in the experimental diet group (control 67 vs experimental diet 45). The difference in the primary end point of the study-sudden death or myocardial infarction was not statistically significant but when these data were pooled with those for cerebral infarction and other secondary end points, the totals were 96 in the control group and 66 in the experimental group; P=0.01. Fatal atherosclerotic events occurred in 70 patients in the control group and 48 in the experimental group (P<0.05). For all primary and secondary end points the incidence rates were 47.7% and 31.3% for the control and experimental groups respectively (P= 0.02).

 

The Finnish Mental Hospital Study was carried out in two mental hospitals. One hospital was switched to a diet low in SFA and cholesterol and relatively high in PUFA, while the other hospital continued the usual hospital diet (46-48). After 6 years the type of diet was reversed in each hospital. The individuals in this study were hospitalized men between 34 to 64 years of age and women age 44 to 64 years. During the study individuals were removed from the study and others added to the study cohort. The serum cholesterol level on the usual diet was 268mg/dL while on the low SFA diet the serum cholesterol level was 226mg/dL. The incidence of CVD was consistently much lower during the low SFA diet periods than during the normal-diet periods but detailed comparisons are difficult due to the lack of randomization of individuals and the adding and removal of individuals during the study leading to only 36% of the men and 20.6% of the women completing both periods of the study. Nevertheless, this study provides evidence of the benefit of a diet low in SFA and cholesterol and enriched in PUFA.

 

The Sydney Diet Heart Study was a randomized controlled trial conducted from 1966 to 1973 that evaluated the effects of increasing linoleic acid from safflower oil (PUFA ~ 15% of calories) in place of SFA (<10% of calories) in men aged 30-59 years with a history of coronary artery disease (49). Participants were randomized to the dietary intervention group (n=221) or a control group with no specific dietary instruction (n=237). Baseline cholesterol levels were ~280mg/dL and decreased to 267mg/dL in the control group and 244mg/dL in the diet intervention group. Compared with the control group, the intervention group had an increased risk of all-cause mortality (17.6% v 11.8%; P=0.051), cardiovascular mortality (17.2% v 11.0%; P=0.037), and mortality from coronary heart disease (16.3% v 10.1%; P=0.036) over the 5 years of the trial. The reason for the increase in mortality is not clear but the safflower oil margarine substitute for animal fats may have contained trans fatty acids, which could have increased CVD.

 

The DART trial was a multicenter trial in men less than 70 years of age with a diagnosis of an acute myocardial infarction (50). There were several different dietary approaches used in this trial but the one of interest reduced fat intake to 30% of total energy and increased the PUFA/SFA ratio to 1.0 (n=1018) vs. no advice (n=1015). The fat advice group reduced SFA from 15% to 11% of total calories, increased PUFA from 7% to 9%, and increased carbohydrate intake from 44% to 46%. Cholesterol levels were reduced by 3.6% (baseline 252mg/dL) in the diet advice group. During the 2-year trial the number of cardiovascular events were similar in the diet group vs. no advice group.

 

The Minnesota Coronary Survey was a 4.5-year, randomized trial that was conducted in six Minnesota state mental hospitals and one nursing home and included 4,393 men and 4,664 women (51). The trial compared the effects of the usual diet (18% SFA, 5% PUFA, 16% monounsaturated fatty acid (MUFA), 446 mg dietary cholesterol per day) versus a low SFA and cholesterol treatment diet (9% SFA, 15% PUFA, 14% MUFA, 166 mg dietary cholesterol per day). The mean duration of time on the diets was 384 days, with 1,568 subjects consuming the diet for over 2 years. The baseline serum cholesterol level was 207 mg/dL, falling to 175 mg/dL in the treatment group and 203 mg/dL in the control group. No differences between the treatment and control groups were observed for cardiovascular events, cardiovascular deaths, or total mortality, perhaps due to the relatively short duration of this study.

 

The Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) with increased consumption of vegetables, fruits, and grains. LDL-C levels were reduced by 3.55 mg/dL in the intervention group while levels of HDL-C and TGs were not significantly different in the intervention vs comparison groups. The dietary intervention did not significantly decrease CVD. In fact, in the women with pre-existing CVD there was an increase in cardiovascular events with diet therapy.

 

Summary of Dietary Randomized Controlled Trials 

 

In reviewing these randomized controlled trials, it appears that the dietary studies that produce a long-term decrease in plasma cholesterol levels resulted in a reduction in cardiovascular events (Oslo Diet-Heart Study, soya-bean trial, Los Angeles Veterans Administration Center, Finnish Mental Hospital Study) while the dietary studies that did not produce a long-term decrease in plasma cholesterol levels failed to demonstrate a reduction in CVD. The baseline plasma cholesterol levels in the positive studies tended to be high and allowed for a robust cholesterol lowering with dietary manipulation. Additionally, as will presented in the next section the greater the reduction in SFA in the diet the greater the decrease in TC and LDL-C levels and many of the positive studies were carried out in an era when the content of SFA in the diet was high. Additionally, studies in non-human primates have also demonstrated that reducing SFA intake reduces atherosclerosis (53,54).

 

These results correspond very nicely with the large number of trials demonstrating that using a variety of different pharmacologic agents that lower plasma cholesterol levels results in a decrease in cardiovascular events (55). In an analysis comparing cholesterol lowering with diet vs. drug therapy it was observed that a similar decrease in cardiovascular events occurred adjusting for the magnitude of cholesterol lowering (56). Thus, it would appear that diets that decrease dietary SFA and thereby lead to a significant decrease in plasma cholesterol levels for an extended period of time have benefits on CVD with the caveat that there is not an increase in other nutrients that will adversely affect other parameters thereby negating the beneficial effects of decreasing SFA. For example, an increase in dietary simple sugars for SFA could lead to an increase in TG levels with negative effects.

 

REVERSAL OF ATHEROSCLEROSIS TRIALS

 

Two studies have examined the effect of decreasing dietary SFA on atherosclerotic lesions.

 

The St Thomas’ Atherosclerosis Regression Study (STARS) determined the effect of decreasing dietary saturated fat in the diet (n=26) vs. usual diet (n=24) in men less than 66 years of age with a plasma cholesterol greater than 234mg/dL referred for coronary angiography to investigate angina pectoris or other findings suggestive of coronary heart disease (57). In the diet group total fat intake was reduced to 27% of dietary energy, saturated fatty acid content to 8-10% of dietary energy, and dietary cholesterol to 100 mg/1000 kcal; omega-6 and omega-3 polyunsaturated fatty acids were increased to 8% of dietary energy, and plant-derived soluble fiber intake was increased to the equivalent of 3-6 g polygalacturonate/1000 kcal. During the trial LDL-C levels were 163mg/dL in the diet intervention group vs.182mg/dL in the usual diet group. Additionally, TGs decreased in the diet intervention group (206mg/dL to 165mg/dl) with no change in TG levels in the usual diet group. After approximately 3 years coronary angiography revealed that the percentage of patients who showed progression of coronary narrowing was significantly reduced by the dietary intervention (usual diet 46% vs, dietary intervention 15%), whereas the percentage who showed an increase in luminal diameter rose significantly (usual diet 4% vs. dietary intervention 38%). While the number of cardiovascular events was small, they were significantly reduced in the dietary intervention group (usual diet 36% vs dietary intervention 11%; p< 0.05). Finally, the improvement in angiographic appearance correlated with LDL-C levels.

 

The Lifestyle Heart Trial was a one year randomized, controlled trial to determine whether lifestyle changes affect coronary atherosclerosis in patients with angiographically documented coronary artery disease (58). Patients were assigned to the lifestyle group (low-fat vegetarian diet, stopping smoking, stress management training, and moderate exercise) (n= 22) or a usual-care control group (n=19). The lifestyle diet contained approximately 10% of calories as fat PUFA/SFA ratio greater than 1), 15-20% protein, and 70-75% predominantly complex carbohydrates. Cholesterol intake was limited to 5 mg/day or less. In the lifestyle group LDL-C decreased from 153mg/dL to 96mg/dl (37% decrease) whereas in the usual care group LDL-C decreased from 168mg/dL to 159mg/dL. Patients in the lifestyle group reported a 91% decrease in the frequency of angina, a 42% decrease in the duration of angina, and a 28% decrease in the severity of angina. In contrast, patients in the usual care group reported a 165% increase in the frequency of angina, a 95% increase in the duration of angina, and a 39% increase in the severity of angina. In the lifestyle group regression of coronary atherosclerosis occurred in 18 of the 22 patients (82%) whereas in the usual care group progression of coronary atherosclerosis occurred in 10 of 19 patients (53%).

 

These two regression trials provide strong support for the results observed in the randomized cardiovascular outcome studies described above i.e., that lowering LDL-C levels by decreasing dietary SFA can reduce atherosclerosis and cardiovascular events.

 

Effect of Dietary Saturated Fatty Acids on Lipid Levels          

 

It should be recognized that when one increases or decreases a particular macronutrient in the diet there needs to be a reciprocal change in another macronutrient to maintain caloric balance.

The effect of substituting PUFA, MUFA, or carbohydrates (CHO) for SFA is shown in table 1. Note that this table shows the effect of replacing 5% of energy from SFA for the indicated dietary component. Thus, going from a diet where 15% of the calories is from SFA to a diet where 10% of the calories is from SFA is estimated to lower LDL-C levels from 6 to 9mg/dL depending on which dietary component replaces the SFA. To keep this decrease in LDL-C in perspective it is estimated that a 40mg/dL decrease in LDL-C induced by statin therapy will result in an approximate 20% decrease in cardiovascular events over a 5 year period of time but the lifetime benefits of a 10 mg/dL decrease in LDL-C due to genetic variants will result in a 16–18% decrease in cardiovascular events (59). The effect on TGs is dependent on the dietary component replacing SFA with CHO resulting in a large increase in TG levels. One should note that there is also a decrease in HDL-C with replacement of SFA (table 2).

 

Table 2. Effect of Decreasing Dietary Saturated Fatty Acids on Lipid Levels

Dietary Component

LDL-C (mg/dL)

TGs (mg/dL)

HDL-C (mg/dL)

PUFA

−9.0    

-2.0

-1.0

MUFA

-6.5

+1.0

-6.0

CHO

-6.0

+9.5

-2.0

PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids; CHO- carbohydrates.

Effects on lipoprotein lipids of replacing 5% of energy from SFA with the 5% of energy from the specified dietary component. Table adapted from references (26,60).

 

SFA in the diet predominantly increases LDL-C levels, predominantly larger, cholesterol-enriched LDL, with modest increases in HDL-C (60,61). As expected, Apo B and apo AI levels also increase (60). These effects are observed in both men and women (60). The effect of a decrease or increase in SFA intake on lipids and lipoproteins is linear with a consistent effect on serum lipids and lipoproteins across a wide range of SFA intakes (60). Of note the effects of decreasing SFA intake was observed even when the SFA intake was already less than 10% of the daily energy intake. Most studies have suggested that replacement of SFA with carbohydrate or unsaturated fat modestly increases Lp(a) but the results have varied from study to study with replacement of SFA with unsaturated fat from particular food sources such as nuts showing no increase in Lp(a) (62).

 

Individual SFA have diverse biological and cholesterol-raising effects with chain length of SFA playing an important role in determining the effect on lipid and lipoprotein levels. The most commonly consumed SFA are palmitic acid (16:0; major source: vegetable oil, dairy, and meat), stearic acid (18:0; meat, dairy, and chocolate), myristic acid (14:0; dairy and tropical oil, particularly coconut oil) and lauric acid (12:0; dairy and tropical oil). A meta-analysis of 60 controlled trials by Mensink et al. reported an increase in LDL-C and HDL-C concentrations by isocaloric replacement of carbohydrates with palmitic, myristic, and lauric acids (63). As expected, apolipoprotein B and A-I also increase (60,64). Myristic and palmitic acids increased LDL-C and HDL-C levels to a similar extent, whereas lauric acid had the largest LDL-C- and HDL-C-raising effect (63,65). Stearic acid did not increase LDL-C levels (63,65).The lack of an association between stearic acid and changes in LDL-C levels has been linked to a slower and/or less efficient absorption as well as desaturation of stearic acid to oleic acid (66). Compared with carbohydrates, an increased intake of lauric, myristic, palmitic or stearic acid lowered TG levels (63,65). For a specific individual many factors including lifestyle factors such as overall dietary composition and physical activity, clinical conditions such as obesity, insulin resistance and hypertriglyceridemia, as well as genetic factors may modify these responses.

 

MECHANISM FOR THE INCREASE IN LDL-C

 

Dietary SFA have been shown to decrease hepatic LDL receptor activity, protein, and mRNA levels and this results in a decrease in the clearance of circulating LDL leading to increased LDL-C levels (67,68). Additionally, the decrease in LDL receptors could result in an increase in the conversion of intermediate density lipoproteins to LDL rather than clearance by the liver (i.e., LDL production is enhanced).

 

SFA have been shown to decrease the formation of cholesterol esters, a reaction catalyzed by the enzyme acyl CoA:cholesterol acyltransferase (ACAT) (68). Free cholesterol in the endoplasmic reticulum is the primary regulator of the activation of sterol receptor binding protein (SREBP), which translocates to the nucleus and enhances the transcription of the LDL receptor (69). Elevated levels of cholesterol in the endoplasmic reticulum prevents the activation of SREBP (69). When free cholesterol is esterified into cholesterol esters it no longer prevents the activation of SREBP and the up-regulation LDL receptor expression. Thus, SFA by decreasing the formation of cholesterol esters and increasing free cholesterol may lead to the down-regulation of LDL receptor expression (68).

 

DIETARY MONOUNSATURATED AND POLYUNSATURATED FATTY ACIDS

 

Olive oil, canola oil, peanut oil, safflower oil, sesame oil, avocados, peanut butter, and many nuts and seeds are major sources of MUFA (table 3). Soybean oil, corn oil, sunflower oil, some nuts and seeds such as walnuts and sunflower seeds, tofu, and soybeans are major sources of PUFA (table 3). Omega-3-fatty acids, eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA, 22:6), are mostly found in fish and other seafood, while another omega-3 fatty acid, alpha-linolenic acid (ALA, 18:3) is found mostly in nuts and seeds such as walnuts, flaxseed, and some vegetable oils such as soybean and canola oils. The body is capable of converting ALA into EPA and DHA but the conversion rates are low.

 

Table 3. Fat Composition of Oils, Lard, Butter, and Margarine

Type of Oil

SFA (%)

MUFA (%)

PUFA (%)

Corn oil

13.6

28.97

57.43

Safflower oil (linoleic)

6.51

15.1

78.4

Canola oil

7.46

64.1

28.49

Almond oil

8.59

73.19

18.22

Olive oil

14.19

74.99

10.82

Soybean oil

16.27

23.69

60.0

Sesame oil

14.85

41.53

43.62

Sunflower oil (linoleic)

10.79

20.42

68.8

Avocado oil

12.1

73.8

14.11

Peanut oil

17.77

48.58

33.65

Palm oil

51.57

38.7

9.73

Coconut oil

91.92

6.16

1.91

Lard

41.1

47.23

11.73

Butter

68.1

27.87

4.0

Margarine (soft)

20

47

33

Margarine (hard)

80

14

6

U.S. Department of Agriculture

 

Effect of Dietary Monounsaturated and Polyunsaturated Fatty Acids on Cardiovascular Disease

 

MONOUNSATURATED FATTY ACIDS

 

Many meta-analyses, but not all, have failed to demonstrate that MUFA intake reduces cardiovascular events (29,33,35,70). However, one meta-analysis and the Nurses’ Health Study and Health Professionals Follow-Up Study, two very large observational studies, found that MUFA when delivered from plant sources was protective but MUFA from other sources was not protective from developing cardiovascular events (71,72).

 

The PREDIMED a randomized controlled outcome trial employing a Mediterranean diet (increased MUFA) reduced the incidence of major CVD (73-75). In this multicenter trial, carried out in Spain, over 7,000 individuals at high risk for developing CVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary composite end point (myocardial infarction, stroke, and death from CVD), which was primarily due to a decrease in strokes. The Mediterranean diet resulted in a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels (76). The changes in lipids were unlikely to account for the beneficial effects of the Mediterranean diet on CVD.

 

The Lyon Diet Heart Study randomized 584 patients who had a myocardial infarction within 6 months to a Mediterranean type diet vs usual diet (77,78). The oils recommended for salads and food preparation were rapeseed and olive oils exclusively. Additionally, they were also supplied with a rapeseed (canola) oil-based margarine. There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal myocardial infarction rate was 4.07 per 100 patient years in the control diet vs.1.24 in the Mediterranean diet; p<0.0001). Lipid levels were similar in both groups in this trial (77).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with cardiovascular disease (79). The Mediterranean diet contained a minimum of 35% of the calories as fat (22% monounsaturated fatty acids, 6% polyunsaturated fatty acids, and <10% saturated fat), 15% proteins, and a maximum of 50% carbohydrates while the low-fat diet contained less than 30% of total fat (<10% saturated fat, 12–14% monounsaturated fatty acids, and 6–8% polyunsaturated fatty acids), 15% protein, and a minimum of 55% carbohydrates. The risk of an ASCVD event was reduced by approximately 25-30% in the Mediterranean diet group. Whether these diets differed in their effects on fasting lipid levels has not been reported.

 

The results of these three randomized trials indicate that a Mediterranean diet enriched in plant MUFA reduce the risk of CVD. It is likely that the beneficial effects of the Mediterranean diet on CVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution. It should be noted that in addition to an increase in MUFA the diet also includes low to moderate red wine consumption, high consumption of whole grains and cereals, low consumption of meat and meat products, increased consumption of fish, and moderate consumption of milk and dairy products. As in many dietary studies it is difficult to change a single variable and therefore the interpretation of which factor or factors account for the benefits is difficult to untangle.

 

POLYUNSATURATED FATTY ACIDS

 

Recent meta-analyses of the effect of PUFA on cardiovascular events in observational studies have demonstrated either no effect or a modestly lower risk of CVD and mortality (80-84). Randomized trials are described in the section on saturated fats and CVD and describe the results of replacing SFA with PUFA. It appears that dietary PUFA has a neutral effect on CVD except in the circumstances where it replaces SFA and results in a sustained decrease in plasma cholesterol levels leading to a decrease in cardiovascular events.

 

OMEGA-3-FATTY ACIDS

 

As discussed in detail in the chapter entitled “Triglyceride  Lowering Drugs” numerous randomized controlled trials of the effect of low dose omega-3-fatty acids (approximately ≤1 gram/day) on CVD have been published and the bulk of the evidence indicates no benefit (85). The effect of pharmacologic doses of omega-3-fatty acids (≥1.8 grams/day) on cardiovascular outcomes is discussed in the chapter entitled “Triglyceride  Lowering Drugs” (85).

 

Effect of Dietary Monounsaturated and Polyunsaturated Fatty Acids on Lipid Levels

 

Table 4 shows the effect of substituting PUFA or MUFA for carbohydrates on LDL-C, HDL-C, and TG levels. Both PUFA and MUFA decrease LDL-C and TGs but PUFA induces a greater decrease (60). Both PUFA and MUFA increase HDL-C levels (60).

 

Table 4. Effect of Decreasing Dietary Carbohydrate on Lipid Levels

Dietary Component

LDL-C (mg/dL)

TGs (mg/dL)

HDL-C (mg/dL)

PUFA

-4.3

-9.2

1.2

MUFA

-1.8

-6.6

1.6

PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids;

Effects on lipoprotein lipids of replacing 5% of energy from carbohydrates with the 5% of energy from the specified dietary component. Table adapted from reference (60).

 

In a meta-analysis of 14 studies no significant differences in TC, LDL-C, or HDL-C  levels were observed when diets high in MUFA or PUFA were compared directly (86). TG levels were modestly but consistently lower on the diets high in PUFA (P = .05) (86).

 

While high dose omega-3-fatty acids (3-4 grams/day) lower TG levels, lower doses (≤1 gram/day) have minimal effects on lipid levels (85).

 

MECHANISM FOR THE DECREASE IN LDL-C

 

Unsaturated fatty acids increase hepatic LDL receptor activity, protein, and mRNA abundance, which will increase the clearance of LDL from the circulation (67,68). Unsaturated fatty acids are a preferred substrate for ACAT and thereby result in an increase in cholesterol ester formation and a decrease in free cholesterol in the liver (68). A decrease in hepatic free cholesterol will result in the up-regulation of LDL receptor expression leading to a decrease in LDL-C levels. PUFA also increase membrane fluidity leading to an increase in the ability of LDL receptors to bind LDL (67). Additionally, the increase in LDL receptors could result in a decrease in the conversion of intermediate density lipoproteins (IDL) to LDL due to increased uptake of IDL by the liver (i.e., LDL production is decreased).

 

DIETARY TRANS FATTY ACIDS

 

The two major sources of dietary trans fatty acids (TFA) are those that occur naturally in meat and dairy products as a result of anaerobic bacterial fermentation in ruminant animals and those formed during the partial hydrogenation of vegetable fat (the fatty acids in vegetable oils have cis double bonds) (87). Partial hydrogenation and the formation of TFA converts the liquid vegetable oil into a solid form at room temperature allowing for ease of use in food products and increased shelf life (87,88). TFA acids were widely used in baked products, packaged snack foods, margarines, and crackers (88). With the recognition of the adverse effects of TFA the use of partial hydrogenated oils in food products has markedly diminished World-wide and in the US is no longer allowed.

 

Effect of Trans Fatty Acids on Cardiovascular Disease

 

A meta-analysis by de Souza and colleagues of 5 studies with 70,864 participants found that the relative risk of coronary heart disease mortality disease was increased with dietary TFA (1.28; p=0.003) (34). Similarly, the relative risk of coronary heart disease was also increased (1.21; p<0,001) (34). Another meta-analysis by Chowdhury and colleagues of 5 studies with 155,270 participants found that the relative risk of coronary events was increased with higher intake of TFA (RR 1.16; CI 1.06-1.27) (33). It has been estimated that a 2 percent increase in energy intake from TFA was associated with a 23 percent increase in the incidence of coronary heart disease (88). Thus, observational studies have consistently demonstrated that an increase in dietary TFA increase the risk of CVD. Clearly it would not be ethical to carry out randomized trials of the effect of TFA acids on CVD.

 

Effect of Trans Fatty Acids on Lipid Levels

 

The effect of replacing SFA, MUFA or PUFA with TFA acids is shown in table 5. TFA increase LDL-C levels and decrease HDL-C levels. Of note TFA increase LDL-C even when substituting for SFA. There appears to be a nearly linear relationship between TFA intake and LDL-C concentration, but this relationship does not seem to exist between TFA intake and HDL-C (89). HDL-C seems to be lowered significantly by TFA only when intake is >2% to 4% of the total energy intake (89). TFA also increases TG and Lp(a) levels (88). Additionally, dietary TFA increases small dense LDL and the increase correlates with the quantity of TFA in the diet (90). 

 

Table 5. Effect on Lipids of Replacing Various Fatty Acids with Trans Fatty Acids

Dietary Component

LDL-C (mg/dL)

HDL-C (mg/dL)

SFA

2.0

-2.0

PUFA

11.5

-1.3

MUFA

9.5

-1.5

SFA- saturated fatty acids; PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids. All results are statistically significant (P<0.05) except the increase in LDL-C with SFA replacement. Effects on lipoprotein lipids of replacing 5% of energy from various fatty acids with 5% of the energy from TFA. Table adapted from reference (88).

 

Replacing carbohydrates with TFA results in an increase in LDL-C and apo B and no change in HDL-C, apo AI, or TG levels (63).

 

RUMINANT TRANS FATTY ACID

 

A key question now that TFA derived from partial hydrogenation of vegetable fat in the diet have been markedly reduced is whether ruminant derived TFA which are present in milk, butter, cheese, and beef have harmful effects similar to industrial created TFA. It is important to note that ruminant derived TFA have a different composition with ruminant TFA being enriched in vaccenic acid, which is the predominant TFA, and conjugated linoleic acid (89,91). Also the quantities of ruminant TFA ingested is much lower than the quantities of industrial TFA ingested (89). In an analysis of a large number of studies of the effect of ruminant and industrial TFA on lipid levels it was observed that the effect of ruminant TFA on LDL-C and HDL-C was similar but slightly less than that of industrial TFA (the difference was not significant) (91). Whether the low quantities of ruminant TFA in the diet will influence the risk of CVD is unknown (89) but a meta-analysis of 4 observational trials did not find a link between ruminant-TFA intake (increments ranging from 0.5 to 1.9 g/day) and the risk of CHD (RR=0.92; CI 0.76-1.11; P=0.36) (92). Another meta-analysis also did not find a link between ruminant TFA and CVD (34). 

 

MECHANISM FOR THE LIPID EFFECTS OF TRANS FATTY ACIDS

 

The mechanism for the increase in LDL-C levels by dietary TFA is thought to be due to decreased LDL-Apo B catabolism without a change in LDL-Apo B production (87,93). The decrease in HDL-C induced by TFA has been attributed to an increase in HDL Apo A-I catabolism without a significant change in HDL apoA-1 production rate (87,93). Additionally, TFA increases CETP activity which could increase the transfer of cholesterol esters from HDL to LDL thereby contributing to the decreased HDL-C levels and increased LDL-C levels (94).

 

DIETARY CHOLESTEROL

 

The primary food sources of dietary cholesterol are egg yolks, shrimp, beef, pork, poultry, cheese, and butter with the top five food sources being eggs and mixed egg dishes, chicken, beef, burgers, and cheese (table 6) (95). In the US the typical cholesterol intake varies from 50 to 400mg per day with a mean of 293 mg/day (348 mg/day for men and 242 mg/day for women) (96).

 

Table 6. Cholesterol Content of Food

Food

mg per 100 grams

Egg

373

Butter

215

Shrimp

125

Cheese

108

Beef

90

Chicken

88

Pork

80

Ice Cream

47

 

Effect of Dietary Cholesterol on Cardiovascular Disease

 

In reviews of prospective observational studies an association between dietary cholesterol and CVD has not been clearly demonstrated with some studies reporting an association and others no association (97,98). Most of these studies did not adjust for the amount and types of fatty acids consumed, which could influence the results as foods containing large amounts of cholesterol are also rich in SFA. Dietary cholesterol was not associated with cardiovascular risk among >80,000 nurses and 43,000 male health care professionals after adjusting for energy intake, PUFA, trans fatty acid, and SFA intake (99,100).

 

Most foods that contain cholesterol also contain significant amounts of SFA. An exception are eggs which contain significant amounts of cholesterol and only small amounts of SFA (95). It is therefore of interest to examine the effect of egg consumption on CVD. In an analysis of 7 cohort studies no association between egg intake and coronary heart disease was observed and egg intake may be associated with a reduced risk of stroke (101). A recent meta-analysis of 23 prospective studies with 1,415,839 individuals and a median follow-up of 12.28 years also found that increased consumption of eggs was not associated with increased risk of CVD (102). Other meta-analyses and reviews have also not demonstrated a consistent link between eggs and CVD (98,103-105). However, a recent very large meta-analysis with 3,601,401 participants with 255,479 events showed that the consumption of 1 additional 50-g egg daily was associated with a very small increase in CVD risk (pooled relative risk, 1.04; 95% CI 1.00-1.08) (106). Thus, eggs have either no effect or a very small effect on CVD that can be seen only in very large studies.

 

There appears to be no randomized studies of the effect of decreasing cholesterol intake on CVD. Do recognize that the studies of decreasing dietary SFA intake described earlier also result in a decrease in cholesterol intake. Thus, at this time there is very limited data linking dietary cholesterol intake with an increased risk of CVD. 

 

Effect of Dietary Cholesterol on Lipid Levels   

 

In a meta-analysis of fifty-five studies with 2,652 subjects the predicted change in LDL-C levels for an increase of 100 mg dietary cholesterol per day adjusted for dietary fatty acids ranged from 1.90mg/dL to 4.58 mg/dL depending upon the model employed (107). An increase of 200mg dietary cholesterol per day increased LDL-C levels from 3.80mg/dL to 6.96mg/dL. It should be noted that the effect of dietary cholesterol levels is greater the higher the LDL-C level (107). For a baseline LDL-C level of 100, 125, 150, and 175 mg/dL the predicted increase in LDL-C for a change in dietary cholesterol of 100mg is 2.7, 3.6, 4.6, and 5.5 mg/dL respectively (107). While the absolute increase is greater if the LDL-C level is higher the percentage increase is similar. Moreover, cholesterol feeding does not alter number of LDL particles – instead it increases the cholesterol content of the LDL particles leading to the formation of large buoyant LDL (108).

 

The effect of dietary cholesterol on HDL-C levels differs in males and females. In men an increase of 100mg of dietary cholesterol results in a 0.30 to 1.44mg/dL decrease in HDL-C levels while in women this results in a 0.50 to 1.61 increase in HDL-C levels (107). Dietary cholesterol does not impact TG or VLDL cholesterol levels (97). 

 

Approximately 15-25% of the population have an increased response to dietary cholesterol with greater increases in LDL-C levels (i.e., sensitive or hyper-responders), while the majority respond minimally (i.e., non-sensitive or hypo-responders) (109). An intake of 100 mg/day dietary cholesterol leads to a 3-4-fold difference in LDL-C concentration between hyper- and hypo-responders (an increase of 2.84 mg/dL vs. 0.76 mg/dL (110). The mechanism for the increase in cholesterol absorption in hyper-responders is unknown. On average 50% (typical range 40-60%) of dietary cholesterol is absorbed but this varies from person to person (111). A high-cholesterol diet leads to significant increases in non-HDL-C levels in insulin-sensitive individuals but not in lean or obese insulin-resistant subjects whereas HDL-C levels increased in all 3 groups (112). The above observations demonstrate the variable response of lipid and lipoprotein levels that can occur in response to dietary manipulations and emphasize how the response of an individual can be variable.

 

MECHANISM FOR THE INCREASE IN LDL-C

 

The increase in LDL-C levels by dietary cholesterol is due to a decrease in hepatic LDL receptors (111). Cholesterol absorbed by the small intestine is packaged into chylomicrons which deliver dietary cholesterol to the liver (111). This increases hepatic cholesterol levels which down-regulates the expression of LDL receptors leading to a decrease in the clearance of LDL from the circulation (111). Additionally, the decrease in LDL receptors could result in an increase in the conversion of intermediate density lipoproteins to LDL rather than clearance by the liver (i.e., LDL production is enhanced).

 

DIETARY CARBOHYDRATES

 

Carbohydrates (CHO) can be divided into high-quality CHO, for example fruits, legumes, vegetables, and whole grains, or low-quality CHO, which include refined grains (such as white bread, white rice, cereal, crackers, and bakery desserts), starches (potatoes), and added sugars (sugar-sweetened beverages, candy). The high-quality CHO are typically enriched in fiber and have a low glycemic index/glycemic load (i.e., are slowly absorbed and thus do not rapidly increase plasma glucose levels). The low-quality CHO have a high glycemic index and load and rapidly increase plasma glucose levels.

 

Effect of Dietary Carbohydrates on Cardiovascular Disease

 

OBSERVATIONAL STUDIES

 

When SFA is replaced by CHO there is no reduction in CVD whereas replacement of SFA with high quality CHO may be beneficial (27,37,38). A study by Jakobsen and colleagues found that replacing SFA with CHO with a low-glycemic index value is associated with a lower risk of myocardial infarction whereas replacing SFA with CHO with a high-glycemic index values is associated with a higher risk of myocardial infarction (113). Meta-analyses and reviews of the association of glycemic index with CVD have varied with some showing an association of low glycemic index with CVD and others reporting no link (114,115). Two very large studies found that a diet with a high glycemic index was associated with an increased risk of cardiovascular disease (116,117). It should be noted that in the largest study the relative risk for CVD was relatively modest (RR 1.15; 95% CI 1.11-1.19) (117). An increase in cardiovascular morbidity and mortality was associated with an increase in added sugar intake (118-121). Hazard ratios were 1.30 (95% CI- 1.09-1.55) and 2.75 (95% CI-1.40-5.42), respectively, comparing participants who consumed 10.0% to 24.9% or 25.0% or more calories from added sugar with those who consumed less than 10.0% of calories from added sugar (118). Additionally, in the Health Professionals Follow-up Study participants in the top quartile of sugar-sweetened beverage intake had a 20% higher relative risk of coronary heart disease than those in the bottom quartile (RR=1.20; 95% CI- 1.09-1.33) after adjustment for multiple risk factors (122).

 

RANDOMIZED CONTROLLED TRIALS

 

Three of the randomized trials described above in the SFA and CVD section provide information on the role of CHO on CVD. The British Medical Research Council studied 252 men after a myocardial infarction aiming to reduce total fat from 41% to 22% of calories and maintaining total fat at 41% in the control group (41). The type of fat was similar in the high- and low-fat groups, mainly saturated fat from dairy products and meat. It is likely that the decrease in fat calories was substituted by an increase in CHO calories. The type of CHO that replaced the SFA was not specified but the authors indicated that there was a marked increase in sugar intake in the low- fat diet group. There was no difference between the two groups in cardiovascular events during the 5 years of the trial. The DART study decreased SFA which were substituted with PUFA and CHO (50). During the 2-year trial cardiovascular events were similar in the decreased SFA vs. PUFA and CHO group. Finally, the Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day (i.e., CHO}. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) fat with increased consumption of CHO. The dietary intervention did not significantly decrease CVD even though the CHO recommended was high quality CHO. These randomized studies do not provide support for a benefit of substituting CHO for fat in reducing CVD. Of particular note is the Women’s Health Initiative which decreased fat intake and increased high quality CHO and observed no cardiovascular benefits in contrast to the results of observational studies.

 

Effect of Dietary Carbohydrates on Lipids

 

Replacing SFA, MUFA, or PUFA with CHO results in an increase in TGs and a decrease in HDL-C levels (60,63). Replacing SFA with CHO results in a decrease in LDL-C while replacing MUFA or PUFA with CHO results in an increase in LDL-C (see tables 2 and 4) (60,63). In addition, dietary CHO increases the quantity of small dense LDL particles (123). The consumption of moderate amounts of fructose or sucrose (40-80 grams/day) in healthy young men was sufficient to increase small dense LDL levels (124). The effect of increasing dietary CHO on Lp(a) levels has been variable (62).

 

Conversely, decreasing CHO in the diet and adding fat results in an increase in LDL-C and HDL-C levels and a decrease in TG levels (125). In a meta-analysis of eleven randomized controlled trials with 1,369 participants comparing low fat/high CHO diet to high fat/low CHO diet it was found that the high fat/low CHO led to an increase in LDL-cholesterol (6.24mg/dL; 95 % CI 0.12- 12.9) and HDL-C (5.46mg/dL; 95% CI 3.51- 7.41) compared with subjects on the low fat/high CHO diets (126). The high fat/low CHO decreased TG levels (-22.9mg/dL; 95 % CI -13.4- -32.6 (126). Another meta-analysis of 23 randomized controlled trials also found that a high fat/low CHO diet increased LDL-C and HDL-C levels and decreased TG levels (127). These studies nicely demonstrates that a high fat diet will increase LDL-C and HDL-C levels while a high CHO diet will increase TG levels and decrease HDL-C levels.

 

COMPARISON OF DIFFERENT CARBOHYDRATES ON LIPIDS

 

A meta-analysis of twenty-eight randomized controlled trials comparing low- with high glycemic index diets (1,272 participants) reported that low glycemic index diets significantly decreased LDL-C levels by 6.2mg/dL; P < 0.0001) with no effect on HDL-C or TGs (128). The decrease in LDL-C was related to the amount of fiber and/or phytosterols in the low glycemic diet (see Fiber and Plant Sterols/Stanols section below).

 

High fructose corn syrup (HFCS) has become a major source of fructose intake (HFCS made for beverages contains 55% fructose and 45% glucose). Because sucrose and HFCS are major contributors to total CHO intake there has been interest in the effect of fructose, glucose, and sucrose on lipid levels. In a comparison of isocalorically substituting starch for glucose, fructose, or sucrose there were no difference in TG levels but there was a decrease in LDL-C (approximately 7.8mg/dL) (129).

 

A meta-analysis by Te Morenga and colleagues examined the effect of the addition of sugar on lipid levels. In studies where energy intake was isocaloric, sugar intake increased TG levels by 11.7mg/dL, LDL-C by 6.6mg/dL, and HDL-C by 0.8mg/dL (130). In a similar meta-analysis by Fattore and colleagues an isocaloric substitution of free sugars for complex CHO increased TGs by 8.3mg/dL, LDL-C by 7.1mg/dL, and HDL-C by 1.3mg/dL (131). The increase in TG and LDL-C levels were larger in the trials where greater amounts of free sugar were employed.

 

In a meta-analysis of adding fructose to the diet there was no significant effect on fasting TG levels at dietary fructose < 100 grams per day but at higher amounts fructose increased fasting TG levels (132). Fructose is more likely to have adverse effects on lipids when intake is high and/or when caloric excess is present. For example, in young healthy individuals, a 2-week intervention with 25% of energy requirements as HFCS or fructose sweetened beverages resulted in significant increases in fasting LDL-C, small dense LDL particles, non-HDL-C, apo B, and HDL-C and postprandial TGs (133). High quantities of glucose did not affect LDL-C, non-HDL-C, Apo B, HDL-C, or postprandial TG levels but did increase fasting TG levels (133).

 

Thus, the effect of CHO on lipids can vary depending upon the particular type of CHO studied (table 7). In the case of glycemic index (complex CHO) and starch vs sugar some of the difference in lipid response could be due to other dietary constituents (i.e., fiber, phytosterols).

 

Table 7. Summary of the Effect of Different Carbohydrates on Lipid and Lipoproteins

Comparisons

Effect on Lipids and Lipoproteins

Low GI vs. High GI

High GI increases LDL-C

Sugar vs. Starch

Sugar increases LDL-C

Sugar vs. Complex CHO

Sugar increases LDL-C and TGs

Fructose vs. Glucose

Fructose increases LDL-C and HDL-C and postprandial TGs

Sugar- sucrose, glucose, or fructose

 

MECHANISM OF THE EFFECTS OF CARBOHYDRATES ON LIPIDS

 

Dietary CHO promote hepatic de novo fatty acid synthesis by providing substrate for fatty acid synthesis (Figure 1). This is particularly the case when there is caloric excess. Additionally, the glucose provided by dietary CHO stimulates insulin secretion which also increases hepatic fatty acid synthesis. The increase in fatty acid synthesis in the liver enhances TG synthesis which promotes VLDL formation and secretion leading to an increase in plasma TG levels. 

 

Figure 1. Carbohydrates stimulate VLDL production by stimulating de novo fatty acid synthesis.

 

Fructose is more potent at increasing de novo fatty acid synthesis than glucose. Small quantities of fructose in the diet are metabolized in the small intestine to glucose and organic acids and do not affect systemic metabolism while high quantities of fructose can escape intestinal metabolism and are delivered to the liver (134). In the liver fructose but not glucose activates SREBP1c and ChREBP leading to the increased expression of the genes that synthesize fatty acids stimulating hepatic lipogenesis (134,135). Additionally, fructose metabolism in the liver is not inhibited providing an unlimited supply of fructose carbons for lipogenesis. In contrast, the first steps in glucose metabolism can be inhibited and thus the utilization of glucose for lipogenesis is regulated (134). In addition, fructose inhibits fatty acid oxidation whereas glucose does not (135). These differences in the metabolism of fructose and glucose in the liver explain the increased ability of fructose to stimulate hepatic lipogenesis and the enhanced formation and secretion of VLDL. In the addition to increased VLDL production fructose does not stimulate the secretion of insulin, which plays a key role in stimulating lipoprotein lipase activity and the clearance of TG rich lipoproteins. The failure of dietary fructose to induce an increase in lipoprotein lipase activity may lead to a decrease in the clearance of TG rich lipoproteins compared to dietary glucose, which stimulates insulin secretion.

 

The elevation in TG rich lipoproteins in turn may have effects on other lipoproteins (25) (Figure 2). Specifically, cholesterol ester transfer protein (CETP) mediates the equimolar exchange of TGs from TG rich VLDL and chylomicrons for cholesterol from LDL and HDL (25). The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content and decreasing the cholesterol content of both LDL and HDL particles. This CETP-mediated exchange underlies the commonly observed reciprocal relationship of low HDL-C levels when TG levels are high and the increase in HDL-C when TG levels decrease. The TG on LDL and HDL are then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL particles.

 

Figure 2. The effect of hypertriglyceridemia on LDL and HDL.

 

DIETARY PROTEIN

 

Effect of Dietary Protein on Cardiovascular Disease

 

In a meta-analysis of 10 studies with 425 ,781 participants intake of plant protein was associated with a decrease in cardiovascular mortality (136). Other meta-analyses have also found that intake of plant proteins was associated with a lower risk of cardiovascular mortality (137-139). In some but not all studies animal protein intake increased the risk of cardiovascular mortality (136-139). The differences in outcomes observed between plant and animal proteins could be due to increased intake of SFA with animal proteins and increased fiber and phytosterol intake with plant proteins. 

 

Effect of Dietary Protein on Lipids

 

Because a high protein diet is often associated with an increase in SFA intake it is important to control for this variable in determining the effect of dietary protein on lipid levels. In a meta-analysis of a high vs. low protein diets in individuals on a low-fat diet no difference in LDL-C, HDL-C, or TG levels were observed (140). In another meta-analysis of 24 trials with 1,063 participants that compared isocaloric diets matched for fat intake but with differences in protein and CHO  intakes no differences in LDL-C and HDL-C levels were observed but TG levels were decreased in the high protein diet group (-20.2mg/dL) (141). Greater weight loss and decreased CHO intake in the high protein diet group likely contributed to the decrease in TGs. In a meta-analysis where fat intake was not controlled the high protein diet was associated with an increase in HDL-C levels and a decrease in TG levels (142). It is obviously difficult to determine the effect of dietary protein on lipid levels as other dietary constituents are changing (SFA, CHO) and secondary effects induced by changes in protein intake (weight loss) could influence lipid levels.

 

DIETARY FIBER

 

Dietary fiber are non-digestible carbohydrates including non-starch polysaccharides, cellulose, pectins, hydrocolloids, fructo-oligosaccharides and lignin. Fiber is found mostly in fruits, vegetables, whole grains, nuts, seeds, psyllium seeds, beans, and legumes. There are two main types of dietary fiber; soluble and insoluble. The main sources of soluble fiber are fruits and vegetables and insoluble fiber are cereals and whole-grain products. Most high fiber foods contain both soluble and insoluble fiber. A summary of the fiber content of some foods is shown in tables 8-11.

 

Table 8. Fiber Content of Selected Vegetables

Vegetables

 

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Cooked vegetables

Turnip

½ cup

4.8

1.7

3.1

Peas, green, frozen

½ cup

4.3

1.3

3.0

Okra, frozen

½ cup

4.1

1.0

3.1

Potato, sweet, flesh

½ cup

4.0

1.8

2.2

Brussels sprouts

½ cup

3.8

2.0

1.8

Asparagus

½ cup

2.8

1.7

1.1

Kale

½ cup

2.5

0.7

1.8

Broccoli

½ cup

2.4

1.2

1.2

Carrots, sliced

½ cup

2.0

1.1

0.9

Green beans, canned

½ cup

2.0

0.5

1.5

Beets, flesh only

½ cup

1.8

0.8

1.0

Tomato sauce

½ cup

1.7

0.8

0.9

Corn, whole, canned

½ cup

1.6

0.2

1.4

Spinach

½ cup

1.6

0.5

1.1

Cauliflower

½ cup

1.0

0.4

0.6

Turnip

½ cup

4.8

1.7

3.1

Raw vegetables

Carrots, fresh

1, 7 ½ in. long

2.3

1.1

1.2

Celery, fresh

1 cup chopped

1.7

0.7

1.0

Onion, fresh

½ cup chopped

1.7

0.9

0.8

Pepper, green, fresh

1 cup chopped

1.7

0.7

1.0

Cabbage, red

1 cup

1.5

0.6

0.9

Tomato, fresh

1 medium

1.0

0.1

0.9

Mushrooms, fresh

1 cup pieces

0.8

0.1

0.7

Cucumber, fresh

1 cup

0.5

0.2

0.3

Lettuce, iceberg

1 cup

0.5

0.1

0.4

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 9. Fiber Content of Selected Legumes

Legumes (cooked)

Serving Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Kidney beans, light red

½ cup

7.9

2

5.9

Navy beans

½ cup

6.5

2.2

4.3

Black beans

½ cup

6.1

2.4

3.7

Pinto beans

½ cup

6.1

1.4

4.7

Lentils

½ cup

5.2

0.6

4.6

Black-eyed peas

½ cup

4.7

0.5

4.2

Chick peas, dried

½ cup

4.3

1.3

3

Lima beans

½ cup

4.3

1.1

3.2

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 10. Fiber Content of Selected Fruits

Fruits

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Apricots, fresh w/skin

4

3.5

1.8

1.7

Raspberries, fresh

1 cup

3.3

0.9

2.4

Figs, dried

1 ½

3

1.4

1.6

Mango, fresh

½ small

2.9

1.7

1.2

Orange, fresh

1 small

2.9

1.8

1.1

Pear, fresh, w/skin

½ large

2.9

1.1

1.8

Apple, red, fresh w/skin

1 small

2.8

1

1.8

Strawberries, fresh

1 ¼ cup

2.8

1.1

1.7

Plum, red, fresh

2 medium

2.4

1.1

1.3

Applesauce, canned

½ cup

2

0.7

1.3

Apricots, dried

7 halves

2

1.1

0.9

Peach, fresh, w/skin

1 medium

2

1

1

Kiwifruit, fresh

1 large

1.7

0.7

1

Prunes, dried

3 medium

1.7

1

0.7

Grapefruit, fresh

½ medium

1.6

1.1

0.5

Blueberries, fresh

¾ cup

1.4

0.3

1.1

Cherries, black, fresh

12 large

1.3

0.6

0.7

Banana, fresh

½ small

1.1

0.3

0.8

Melon, cantaloupe

1 cup cubed

1.1

0.3

0.8

Watermelon

1 ¼ cup cubed

0.6

0.4

0.2

Grapes, fresh w/skin

15 small

0.5

0.2

0.3

Raisins, dried

2 tbsp

0.4

0.2

0.2

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 11. Fiber Content of Grains

Food

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Wheat bran

½ cup

12.3

1.0

2.7

Barley, pearled, cooked

½ cup

3.0

0.8

2.2

Oatmeal, dry

⅓ cup

2.7

1.4

11.3

Bread, pumpernickel

1 slice

2.7

1.2

1.5

Wheat flakes

¾ cup

2.3

0.4

1.9

Bread, rye

1 slice

1.8

0.8

1.0

Bread, whole wheat

1 slice

1.5

0.3

1.2

Rice, white, cooked

½ cup

0.8

trace

0.8

Bread, white

1 slice

0.6

0.3

0.3

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Effect of Dietary Fiber on Cardiovascular Disease

 

Several meta-analyses have demonstrated that an increase in total fiber, soluble fiber, and insoluble fiber are associated with a decrease in cardiovascular events (143-148). The greater the intake of fiber the greater the reduction in risk of cardiovascular events.  

 

Effect of Dietary Fiber on Lipids

 

In a meta-analysis of randomized controlled trials the effect of fiber on lipid levels was evaluated (149). Increased dietary fiber decreased total cholesterol (TC) (−7.8mg/dL; 95% CI −13.3 to −2.3), LDL-C (−5.5mg/dL; 95% CI −8.6 to −2.3), and HDL-C levels ( −1.17mg/dL; 95% CI −2.34 to −0.39) (149,150), There was no change in TG levels. A meta-analysis of randomized controlled studies of whole-grain foods vs non-whole-grain foods found that the whole-grain diet lowered LDL-C (-3.51mg/dL; P < 0.01) and TC levels (-4.68mg/dL; P < 0.001) compared with the non-whole grain foods (151). HDL-C and TG levels were not significantly altered by the whole grain diet. Moreover, 3.4 g of psyllium (Metamucil), a soluble fiber, decreased LDL-C with no significant effects on HDL-C or TGs (152,153). In a meta-analysis of 28 randomized trials psyllium lowered LDL by 12.9mg/dL (P < 0.00001) (154). A mean reduction in LDL-C concentrations of about 1.1 mg/dL can be expected for each g of water-soluble fiber in the diet (155,156).

 

MECHANISM OF EFFECT OF FIBER ON LDL-C

 

Fiber is thought to decrease cholesterol absorption by the small intestine (157,158). This leads to a decrease in cholesterol content of chylomicrons and a reduction in the delivery of cholesterol to the liver. The decrease in cholesterol in the liver upregulates LDL receptors resulting in a decrease in plasma LDL-C levels. Fiber may also decrease small intestinal absorption of bile acids which will lead to the increased utilization of hepatic cholesterol for the synthesis of bile acids (159). This will also decrease hepatic cholesterol levels inducing an increase in the expression of LDL receptors lowering plasma LDL-C levels. Finally, colonic fermentation of dietary fiber with production of short-chain fatty acids, such as acetate, propionate, and butyrate, is postulated to inhibit hepatic cholesterol synthesis contributing to a decrease in LDL-C levels (159). 

 

PLANT STEROLS AND STANOLS (PHYTOSTEROLS)

 

Plant sterols and plant stanols (phytosterols) are naturally occurring constituents of plants and are found in vegetable oils, such as corn oil, soybean oil, and rapeseed oil and cereals, nuts, fruits, and vegetables. The intake of plant sterols and stanols is about 200–400 mg/day. The most commonly occurring phytosterols in the human diet are β-sitosterol, campesterol, and stigmasterol. Higher intakes can be achieved by consuming a vegetable-based diets such as a vegetarian diet (400-800mg/day) or by consuming food products enriched with plant sterols or stanols (for example margarines or yogurt). If using foods enriched in phytosterols it is best to take them with main meals to enhance their effectiveness. High doses of phytosterols can affect the absorption of fat-soluble vitamins. The plant sterol and stanol content of different foods is shown in table 12.

 

Table 12. Plant Sterol and Stanol Contents in Different Foods

Food item

Plant Sterols

(mg/100 g)

Plant Stanols

(mg/100 g)

Vegetable oils

Corn oil

686-952

23-33

Rapeseed oil (canola oil)

250-767

2-12

Soybean oil

221-328

7

Sunflower oil

263-376

4

Olive oil

144-193

0.3-4

Palm oil

60-78

Traces

Cereals

Corn

66-178

-

Rye

71-113

12-22

Wheat

45-83

17

Barley

80

2

Millet

77

-

Rice

72

3

Oats

35-61

1

Vegetables

Broccoli

39

2

Cauliflower

18-40

Traces

Carrot

12-16

Traces

Lettuce

9-17

0.5

Potato

7

0.6

Tomato

7

1

Fruits and berries

Avocado

75

0.5

Passion fruit

44

Not detected

Raspberry

27

0.2

Orange

24

Not detected

Apple

12-18

0.8

Banana

12-16

Not detected

Adapted from Piironen V and Lampi AM (160)

 

Effect of Phytosterols on Cardiovascular Disease

 

There is minimal data on the effect of phytosterols on cardiovascular events. From the effect on LDL-C levels one would anticipate that phytosterols would reduce CVD.

 

Effect of Phytosterols on Lipids

 

Plant sterols or plant stanols at a dose of 3 grams per day lowers LDL-C by approximately 12% (161). Higher doses do not dramatically further lower LDL-C levels and lower doses have less effect on LDL-C (for example 2 grams/day lowers LDL-C by 8%) (161).  HDL-C levels are not affected by plant sterols or stanols but TG levels decrease modestly (~6%) with a greater absolute reduction in individuals with high TG level (percent change is the same) (162). To achieve these high doses consuming food products enriched is phytosterols is necessary.

 

MECHANISM OF EFFECT OF PHYTOSTEROLS ON LDL-C

 

Plant sterols or plant stanols reduce LDL-C levels by competing with cholesterol for incorporation into micelles in the gastrointestinal tract, resulting in decreased cholesterol absorption (163). This leads to the decreased delivery of cholesterol to the liver and the up-regulation of LDL-receptor expression lowering LDL-C levels.

 

SUMMARY OF THE EFFECT OF DIETARY CONSTITUENTS ON LIPID LEVELS

 

A summary of the major effects of dietary constituents on lipid levels is shown in table 13, typically under isocaloric feeding conditions in short-term feeding studies. Dietary SFA, TFA, and cholesterol increase LDL-C levels whereas CHO increases TG levels. MUFA, PUFA, fiber and phytosterols decrease LDL-C and TFA decrease HDL-C levels.

 

Table 13. Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

CHO

Increase TGs, increase greater with simple sugars particularly fructose

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

 

EFFECT OF SPECIFIC FOODS ON CARDIOVASCULAR DISEASE

 

There are a large number of observational trials linking various foods with either an increased or decreased risk of CVD. A large meta-analysis by Micha et al reported that fruits, vegetables, beans/legumes, nuts/seeds, whole grains, fish, yogurt, fiber, seafood omega-3 fatty acids, polyunsaturated fats, and potassium were associated with a decreased risk of CVD while unprocessed red meats, processed meats, sugar-sweetened beverages, and sodium were associated with an increased risk of CVD (164). A similar meta-analysis by Bechthold et al found that whole grains, vegetables and fruits, nuts, and fish consumption were associated with a decrease in CVD while red meat, processed meat, and sugar sweetened beverage consumption was associated with an increase in CVD (165). Note, as discussed in the introduction, observational studies have limitations and cannot be assumed to indicate cause and effect. Additional one can find other meta-analyses that reach different conclusions than the results described above. For example, a meta-analysis by Zeraatkar et al and a meta-analysis by Vernooij et al reached the conclusion that meat and processed meat were not associated with a significant increase in CVD (20,166). Thus, one needs recognize that while these studies can suggest beneficial and harmful effects of eating certain foods more definitive studies are required to be certain. For a detailed analysis of the limitations of observational dietary studies see articles by Ioannidis and Nissen (1,2).

 

Only a single randomized trial has examined the effect of specific foods on CVD events. The DART trial randomized men with an acute myocardial infarction to at least two weekly portions (200-400 g) of fatty fish (mackerel, herring, kipper, pilchard, sardine, salmon, or trout) (n=1015) or no dietary advice (n=1018) (50). After approximately 2 years total mortality was significantly lower (RR 0.71; CI 0.54-0.93) in the fish advice group than in the no fish advice group, due to a reduction in ischemic heart disease deaths. There were no significant differences in ischemic heart disease events (RR 0.84; CI 0.66-1.07). In a separate portion of the DART trial there was also a group of men with an acute myocardial infarction randomized to increased intake of cereal fiber (18 grams/day) (n=1017) vs. no dietary advice (n=1016). No reduction in cardiovascular events was seen in the cereal fiber group.

 

Clearly addition randomized trials are required to determine the true benefits of specific foods on cardiovascular events.

 

EFFECT OF SPECIFIC FOODS ON LIPID LEVELS

 

In contrast to the paucity of randomized controlled trials on the effect of specific foods on cardiovascular disease there are an abundance of studies on the effect of specific foods on lipid and lipoprotein levels. Given the large number of studies in many instances I will cite the results of meta-analyses to provide the reader with the typical effects that are observed. It should be noted that the effect of specific foods on lipid and lipoprotein levels tend to be small and therefore the results can be inconsistent from study to study.

 

Nuts and Seeds

 

The most consumed edible tree nuts are almonds, hazelnuts, walnuts, pistachios, pine nuts, cashews, pecans, macadamias, and Brazil nuts. Peanuts are botanically groundnuts or legumes, and are widely considered to be part of the nut food group. Nuts are generally consumed as snacks (fresh or roasted), in spreads (peanut butter, almond paste), or as oils or baked goods. Seeds come in all different sizes, shapes and colors. Popular seeds include flax, pumpkin, sunflower, chia, sesame, and mustard seeds.

 

Nuts and seeds are rich in MUFAs, such as oleic acid and in PUFAs, such as linoleic acid and alpha-linolenic acid (ALA). They also contain small amounts of SFA. Almonds, cashews, hazelnuts, pistachios and macadamian nuts have a high MUFA content (>50%) content when compared with other nuts. For other nuts (e.g., Brazil nuts, pine nuts, and walnuts) the PUFA content is high (>50%), while peanuts and pecans have been found to contain relatively high levels of both MUFA and PUFA (table 14). Nuts are a good source of dietary fiber, ranging from 4-11 g/100 g and phytosterols.

 

Table 14. Nutrient Composition of Nuts

Nuts

 

PUFA

(g/100 g)

MUFA

(g/100 g)

SFA

(g/100 g)

Fiber

(g/100 g)

Walnuts

47.2

8.9

6.1

6.7

Peanuts

15.6

24.4

6.3

8.8

Pistachios

13.7

23.8

5.6

10.3

Almonds

12.3

31.6

3.8

12.5

Hazelnuts

7.9

45.7

4.5

9.7

Cashews

7.8

23.8

7.8

3.3

Pecans

21.6

40.8

6.2

9.6

Macadamias

1.5

58.9

12.1

8.6

 

Consumption of nuts and seeds lower TC and LDL-C levels in healthy subjects or patients with moderate hypercholesterolemia (167-172). Nuts had no significant or minimal effect on increasing HDL-C. The benefits of nuts and seeds vary depending on the type, nutrient composition, and quantity of nuts and seeds consumed. Studies have noted that the estimated cholesterol lowering effect of nuts was greater in individuals with higher initial values of LDL-C and in those with a lower baseline BMI (169).

 

Walnuts: A meta-analysis on the effect of walnuts on lipid levels that included 365 participants showed a decrease in LDL-C (9.2 mg/dL), while HDL-C or TG were not significantly affected (173). In another meta-analysis that analyzed 1,059 participants with a walnut enriched diet LDL-C was lowered by 5.5 mg/dL (174).

 

Almonds: A meta-analysis of 15 studies with 534 participants found that almonds decreased LDL cholesterol (5.8 mg/dL; 95% CI: -9.91, -1.75 mg/dL) and apo B (6.67 mg/dL; 95% CI: -12.63, -0.72 mg/dL) (175). Triglycerides, apo A1, and lipoprotein (a) showed no differences.

 

Pistachio nuts: A meta-analysis of twelve randomized studies reported that pistachio nuts decreased LDL-C -3.82 mg/dL (95% CI, -5.49 to -2.16) and TG -11.19 mg/dL (95% CI, -14.21 to -8.17) levels without effecting HDL-C levels (176).

 

A meta-analysis by Houston et al analyzed the effect of a variety of different nuts on lipid levels (table 15) (177). They found that in general nuts lowered LDL-C and minimally lowered TG levels but had no effect on HDL-C levels. A meta-analysis found that whole flaxseed reduced TC and LDL-C by 6 and 8 mg/dL, respectively (178). Thus, both nuts and seeds lower LDL-C levels.

 

Table 15. Effect of Nuts on Lipid Levels

 

Number of analyses

Number of participants

Effect estimate (mmol/L)

95% CI

LDL Cholesterol

Almond

32

2439

-0.15 [-0.22, -0.08]

Brazil nut

4

307

-0.30 [-0.70, 0.11]

Cashew nut

3

432

 0.02 [-0.12, 0.16]

Hazelnut

6

374

-0.01 [-0.15, 0.12]

Macadamia

6

410

-0.11 [-0.27, 0.04]

Mixed nuts

10

791

 0.04 [-0.06, 0.14]

Peanut

10

1021

 0.08 [-0.04, 0.20]

Pecan

6

295

-0.23 [-0.46, 0.00]

Pistachio

12

736

-0.15 [-0.30, 0.00]

Walnut

35

2582

-0.12 [-0.18, -0.06]

Triglycerides

Almond

32

2439

-0.02 [-0.05, 0.02]

Brazil nut

4

307

 0.04 [-0.54, 0.63]

Cashew nut

3

432

-0.02 [-0.11, 0.07]

Hazelnut

5

313

 0.11 [-0.02, 0.25]

Macadamia

5

342

-0.10 [-0.21, 0.00]

Mixed nuts

11

888

-0.01 [-0.07, 0.06]

Peanut

10

1021

-0.09 [-0.16, -0.02]

Pecan

6

295

-0.11 [-0.24, 0.03]

Pistachio

9

498

-0.12 [-0.21, -0.03]

Walnut

35

3109

-0.09 [-0.12, -0.06]

Table based on data from a meta-analysis by Houston et al (177). To convert mmol/L cholesterol to mg/dL multiply by 39 and to convert mmol/L triglycerides to mg/dL multiply by 88.

 

Whole Grains

 

Whole grains include barley, brown rice, buckwheat, bulgur (cracked wheat), millet, oatmeal, and wild rice. Whole grains contain ~80% more dietary fiber than refined grains, as the latter are milled, a process that removes bran and germ. Refined grains include white flour, white rice, white bread, and corn flower. Health benefits ascribed to whole grains are mainly due to the presence of fiber and bran. A meta-analysis of fifty-five trials with 3900 participants comparing various grains found that oat bran was the most effective intervention strategy for lowering LDL-C (- 12.5mg/dL; 95% CI – 17.2 to – 7.4mg/dL) compared with control (179). Oats also reduced LDC (- 6.6mg/dL; 95% CI – 10.9 to 2.73mg/dL). Barley, brown rice, wheat and wheat bran were not effective in improving blood lipid levels compared with controls. Another meta-analysis also found that whole-grain oats decreased LDL-C levels (–16.7 mg/dL; P < 0.0001) (180).

 

Soy Protein

 

Soybeans and soy products as well as supplements contain soy proteins. In a meta-analysis of 43 randomized studies with 2,607 participants the decrease in LDL-C levels reductions for soy protein ranged between −4.2 and −6.7 mg/dL (P<0.006) (181). Numerous other meta-analyses have reported similar decreases in LDL-C (182-187).  In addition, soy protein also decreases TG levels (~2-10mg/dL) and increases HDL-C levels (~1-2mg/dL). Soy protein does not affect Lp(a) levels (188). The amount of soy protein that is recommended for lipid lowering is 25–50 grams per day (189).

 

The decrease in LDL-C is due to the indirect effect of soy protein decreasing the intake of animal protein (SFA and cholesterol) and the intrinsic effects of bioactive compounds in soy protein (190). The intrinsic effect of soy protein might be mediated by phyto-estrogens that could increase levels of HDL-C and TG and decrease levels of LDL-C (189).   

 

Garlic

 

Garlic supplements are available in several different forms, including garlic powder, allicin, aged garlic extract, and garlic oil. Several meta-analyses have shown that garlic lowers TC levels with variable effects on LDL-C, HDL-C, and TG (191-198). Some studies find a decrease in LDL-C and others a decrease in TG levels. The longer the duration of treatment and the higher the baseline TC the greater the effect. In one meta-analysis TC was reduced by 17 ± 6 mg/dL and low-density lipoprotein cholesterol by 9 ± 6 mg/dL in individuals with elevated TC levels (>200 mg/dL) if treated for longer than 2 months (191). In another meta-analysis garlic powder and aged garlic extract were more effective in reducing TC levels, while garlic oil was more effective in lowering serum TG levels (192). In a meta-analysis of garlic administration to patients with diabetes TC decreased 16.9mg/dL, LDL decreased 9.7mg/dL, TG decreased 12.4mg/dL, and HDL-C increased 3.19mg/dL (all p=0.001) (199). Lp(a) levels are not altered by garlic (198).The mechanism by which garlic alters lipid levels is unknown.

 

Tea

 

Green tea contains many catechins (e.g., epigallocatechin-3-gallate) that influence lipid metabolism in animal models and have been shown to upregulate LDL receptors in liver and suppress PCSK9 production (200,201). Epigallocatechin gallate may also interfere with the intestinal absorption of lipids (202). Most but not all meta-analyses have shown that drinking green tea or black tea decreases TC and LDL-C levels with no significant effect on HDL-C or TG levels (203-214). The reduction in LDL-C is approximately 5-10mg/dL.

 

Coffee

 

Coffee contains cholesterol-increasing compounds; diterpenes such as cafestol and kahweol (215,216). The amount of these cholesterol increasing compounds in coffee depends on how the coffee is prepared (215,216). Boiling coffee beans extracts diterpenes due to the prolonged contact with hot water resulting in high concentrations in the coffee whereas brewed filtered coffee because of the short contact with hot water and retention of diterpenes by the filter paper has lower concentrations of diterpenes. Instant coffee has very low levels of diterpenes (216). The concentration of the cholesterol-raising compound cafestol is negligible in drip-filtered, instant, and percolator coffee but high in unfiltered coffee such as French press, Turkish, or Scandinavian boiled coffee. Levels of cafestol are intermediate in espresso and coffee made in a Moka pot.

 

A meta-analysis of 18 trials found that the consumption of unfiltered, boiled coffee dose-dependently increased TC and LDL-C concentrations (23 mg/dL and 14 mg/dL, respectively), while consumption of filtered coffee resulted in only small changes (TC increased by 3 mg/dL and no effect on LDL-C concentration) (217). Additionally, decaffeinated coffee had a smaller effect and the increase in cholesterol levels was greatest in individuals with hypercholesterolemia. Thus coffee, depending upon how it is prepared, can increase TC and LDL-C levels.

 

Chocolate and Cocoa

 

Cocoa is the non-fat component of finely ground cocoa beans that is used to produce chocolate. Cocoa is rich in flavanols which are low‐molecular‐weight monomeric compounds, such as epicatechin or complex higher‐molecular‐weight oligomeric and polymeric compounds (218). The flavanol content in cocoa products can vary greatly and is dependent on the crop type, post‐harvest handling practices, and manufacturer processing techniques. The flavanol content of milk and white chocolate is low or even absent (218).

 

In a meta-analysis of 21 studies with 986 participants very small effects on LDL-C and HDL-C levels were observed (LDL-C 2.7mg/dL decrease; HDL-C 1.2mg/dL increase) with no change in TG levels with chocolate and/or cocoa intake (219). In another meta-analysis there was a decrease in TG levels (-8.8mg/dL), an increase in HDL-C (2.3mg/dL), and a non-significant decrease in LDL-C (-10.1mg/dL) (220). In studies where the epicatechin dose was greater than 100mg per day the decrease in LDL-C levels was greater (5.5mg/dL) (219). Another meta-analysis of 19 studies found that LDL decreased by 3.3mg/dL and HDL-C increased by 1.8mg/dL with cocoa intake (221). A meta-analysis of 10 clinical trials with 320 participants that focused on dark chocolate found a 6.23mg/dl decrease in LDL-C with no significant changes in HDL-C and TG (222). Thus chocolate/cocoa causes a small decrease in LDL-C levels. 

 

Alcohol

 

It is recommended that females consume no more than 1 drink per day of alcohol (equivalent to 15 grams per day) and that males consume no more than 2 drinks per day (equivalent to 30 grams per day). Alcohol has a relatively high caloric level (7 calories/gram).

 

EFFECT OF ALCOHOL ON LIPID LEVELS

 

In a meta-analysis of 25 studies with an average consumption of 40.9 grams of alcohol per day HDL-C concentrations increased by 5.1 mg/dL (223). HDL-C levels increased by 0.122- 0.133 mg/dL per gram of alcohol per day. Consuming 30 grams of alcohol a day would therefore increase HDL-C concentrations by approximately 3.99 mg/dL compared with an individual who abstains (an 8.3% increase from pretreatment values). The increase in HDL-C was observed regardless of sex, duration of study, median age, or beverage type but the increase was greater in individuals with baseline HDL-C < 40mg/dL and who were sedentary. As expected apo A1 levels also increased. In a meta-analysis of 35 studies TG concentrations increased by 0.19 mg/dL per gram of alcohol consumed a day (P=0.001) and 5.69 mg/dL per 30 g consumed per day (5.9% increase over baseline) (223). The increase in TG levels was seen regardless of beverage type and appeared to be greater in males than females.

 

In a more recent meta-analysis of 33 studies with 796 participants HDL-C levels were increased by 3.67mg/dL by alcohol intake (224). Apo A1 levels were also increased but there were no significant differences in TC, LDL-C, TG, or Lp(a) with alcohol intake. The greater the consumption of alcohol the greater the increase in HDL-C levels. When the consumption of alcohol was greater than 60 grams per day (4 drinks) TG levels were also increased (24.4mg/dL).

 

In a meta-analysis of 14 studies, comparing 548 beer drinkers and 532 controls TC levels were significantly higher in the beer drinkers compared to controls (difference 3.52 mg/dL; p<0.001) (225). In a meta-analysis of 18 studies, comparing 626 beer drinkers and 635 controls HDL-C levels were higher in the beer drinkers compared to controls (difference 3.63 mg/dL: p<0.001) (225). This increase in HDL-C levels in beer drinkers were seen in both males and females. LDL-C and TG levels were not significantly different between beer drinkers and controls (LDL-C difference -2.85 mg/dL; p = 0.070; TG difference 0.40 mg/dL; p = 0.089) (225).

 

Genetic factors play a role in the HDL response to alcohol (226). Individuals with an apoE2 allele have greater HDL-C increase and those with an apoE4 allele have a blunted increase in HDL-C with alcohol intake (226).In addition to an increase in HDL-C levels studies have suggested that the ability of HDL to facilitate the efflux of cholesterol from cells is enhanced by alcohol intake (226,227).

 

One should note in the meta-analyses described above alcohol doesn’t appear to have a major impact on TG levels. However, it must be recognized that the amount of alcohol consumed is a key variable (228,229). At low to moderate amounts alcohol has either no effect or might even decrease TG levels (228). However, at high amounts of alcohol intake increases in TG levels are observed (228,229). As noted above one meta-analysis noted that the consumption of 60 grams per day of alcohol increased TG levels (224). Moreover, alcohol consumed with a meal increases and prolongs the postprandial increase in TG levels (228,229). Additionally, genetic factors and the presence of other abnormalities play a role in the TG response to alcohol intake (229). For example, the increase in TG levels after red wine was -4%, 17%, and 33% in individuals with a BMI 19.60-24.45, 24.46- 26.29, and 26.30-30.44, respectively (P = .001) demonstrating that the increase in TG was strongly influenced by BMI (230). Finally, in patients with pre-existing hypertriglyceridemia moderate alcohol intake increased TG levels (338 ± 71 mg/dL to 498 ± 117 mg/dL; P < 0.05) (231).  

 

MECHANISM FOR THE EFFECT OF ALCOHOL ON HDL

 

 The mechanism for the increase in HDL-C levels is likely due to an increased production of apo A1 and A2 (232). Additionally, alcohol inhibits cholesteryl ester transfer protein (CETP) activity, which will also increase HDL-C levels (229).

 

MECHANISM FOR THE EFFECT OF ALCOHOL ON TRIGLYCERIDES

 

Alcohol increases VLDL secretion by the liver (229). The increased production and secretion of VLDL is due to a number of factors including a) alcohol increases lipolysis in adipose tissue and increases the delivery of fatty acids to the liver b) alcohol increases hepatic fatty acid transporters increasing the uptake of circulating fatty acids c) alcohol increases hepatic de novo fatty acid synthesis d) alcohol decreases the beta oxidation of fatty acids in the liver (229,233,234). Together these effects lead to an increased supply of fatty acids in the liver facilitating TG synthesis and the formation and secretion of VLDL.

 

While moderate alcohol intake increases lipoprotein lipase (LPL) activity acute alcohol intake inhibits LPL activity, which could explain the observation that alcohol consumed with a meal increases postprandial TG levels ((228,229).

 

Summary of the Effect of Specific Foods on Lipid Levels

 

Table 16. Major Effects of Specific Foods on Lipid Levels

Nuts and Seeds

Decrease TC and LDL-C

Whole Grains

Decrease LDL-C

Garlic

Decrease TC, LDL-C, TG

Green and Black Tea

Decrease TC and LDL-C

Coffee (depends on method of preparation)

Increase TC, LDL-C, TG

Cocoa and Chocolate

Decrease LDL-C

 

SPECIFIC DIETS

 

The effect of several dietary strategies on lipid levels is discussed below. Randomized controlled trials on the effect of specific diets on cardiovascular outcomes were discussed in earlier sections (saturated fatty acids section and monounsaturated fatty acids section).  

 

Mediterranean Diet

 

Mediterranean diets have an abundance of plant foods, including vegetables, legumes, nuts, fruits, and grains, fish, and low to moderate red wine consumption. Low consumption of meat and meat products and moderate consumption of milk and dairy products is encouraged. In the PREDIMED trial the Mediterranean diet resulted in a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels (76). In the Lyon Diet Heart Study lipid levels were similar in the Mediterranean and usual diet groups (77). The cardiovascular outcome benefits of both of these randomized outcome trials are discussed in the effect of MUFA on CVD section. In a meta-analysis of the effect of a Mediterranean diet on lipid levels little or no change in LDL-C, HDL-C, and TGs was observed (235). Another meta-analysis reported a 4.6mg/dL decrease in LDL-C and a 0.61mg/dL increase in HDL-C (236).

 

Dietary Approach to Stop Hypertension (DASH) Diet

 

The DASH diet promotes the consumption of fruits, vegetables, low-fat dairy products, whole grains, poultry, fish, and nuts and a decrease in the intake of red meat, sweets, sugar-containing beverages, total fat, saturated fat, and cholesterol. In the initial DASH trial total fat and SFA intake was reduced in the DASH diet group (total fat 27% vs. 39% of calories; SFA 6.2% vs. 15% of calories). MUFA and PUFA intake were similar but cholesterol intake was decreased (194mg/day vs 324mg/day). As expected, CHO and fiber intake were increase (CHO 59% vs. 49% of calories; fiber 35grams/day vs. 17grams/day). The DASH diet lowered TC (15.6 to 19.5mg/dL), LDL-C (11.7 to 15.5mg/dL), and HDL-C (3.12 to 3.90mg/dL) (237). TG levels were not significantly affected. In a meta-analysis of twenty studies of the DASH diet reporting data for 1917 participants TC was decreased (7.8mg/dL; P=0.001) and LDL was decreased (3.9mg/dL; p<0.03) (238). HDL-C and TG levels were not significantly altered (238). Similar results were seen in other meta-analyses (239,240).

 

Portfolio Diet

 

The portfolio dietary pattern is a plant-based dietary pattern that includes four cholesterol-lowering foods; a) tree nuts or peanuts, b) plant protein from soy products, beans, peas, chickpeas, or lentils, c) viscous soluble fiber from oats, barley, psyllium, eggplant, okra, apples, oranges, or berries, and d) plant sterols initially provided in a plant sterol-enriched margarine. In a meta-analysis of 5 studies with 439 participants LDL-C was lowered by 17% (28.5mg/dL; p< 0.001) and TGs by 16% (24.6mg/dL; p< 0.001) with no change in HDL-C or weight (241).

 

Nordic Diet

 

The Nordic diet is based on the consumption of different healthy foods such as whole grains, fruits (such as berries, apples, and pears), vegetables, legumes (such as oats, barley, and almonds), rapeseed oil, fatty fish (such as salmon, herring and mackerel), shellfish, seaweed, low-fat choices of meat (such as poultry and game), low-fat dairy, and decreased intake of salt and sugar-sweetened products. In a meta-analysis of 5 studies LDL-C was decreased by 11.7mg/dL (p= 0.013) with no changes in TG or HDL-C levels (242). In another meta-analysis of 6 studies LDL-C was decreased by 10.1mg/dL with no changes in TG or HDL-C levels (243).

 

Ketogenic Diet

 

Low CHO diets can contain variable amounts of CHO. When the CHO levels are very low, they stimulate the formation of ketones. In a typical ketogenic diet CHO contribute <10% of calories (< 50 grams/day), protein approximately 30% of calories, and fat approximately 60% of calories with no restrictions on the type of fat or cholesterol levels. These diets can be high in beef, poultry, fish, oils, various nuts/seeds, and peanut butter, with moderate amounts of vegetables, salads with low-carbohydrate dressing, cheese, and eggs. Fruits and fruit juices, most dairy products with the exception of hard cheeses and heavy cream, breads, cereals, beans, rice, desserts/sweets, or any other foods containing substantial amounts of CHO are avoided.

 

It is well recognized that a ketogenic diet results in an increase in LDL-C levels, which varies depending upon the type of fat ingested, the degree of carbohydrate restriction, the presence of other medical conditions, weight loss on the diet, and genetic background (244). This increase in LDL-C levels is best illustrated in children treated with a ketogenic diet for epilepsy and in healthy individuals on a ketogenic diet (245-250). In some studies HDL-C is also increased (246-249). A meta-analysis of randomized studies in normal-weight adults found that a ketogenic diet increased LDL-C by 42mg/dL and HDL-C by 13.7mg/dL with no significant changes in TG levels (251). It should be noted that the increase in LDL-C is often not observed or is modest in patients with obesity or the metabolic syndrome (252,253).

 

While the typical increases in LDL-C levels observed with a ketogenic diet are relatively modest, recently a series of reports have described marked elevations in LDL-C levels in some patients on a ketogenic diet (253-255). For example, Goldberg et al reported 5 patients with marked increases in LDL-C levels on a ketogenic diet (256). Three patients had LDL-C levels greater than 500mg/dL. Similarly, Schaffer et al described 3 patients in which a very low carbohydrate diet induced LDL-C levels greater then 400mg/dL (257). Finally, Schmidt et al reported 17 patients with LDL-C levels greater than 200mg/dL on a ketogenic diet (258). In these patients there was an average increase in their LDL-C level of 187 mg/dL (258). The elevations in LDL-C levels decrease towards normal with cessation of the ketogenic diet (256-258). It should be noted that most of the patients with marked elevations in LDL-C in response to a ketogenic diet had normal LDL-C levels prior to the dietary change (255).

 

Many of the individuals who develop marked increases in LDL-C on a very low carbohydrate ketogenic diet have low TG levels, elevated HDL-C levels, and are thin (253,255). This phenotype has been called the lean mass hyper-responder (LMHR) phenotype (253,255). LMHR individuals have been defined as having TG <70mg/dL, HDL-C > 80mg/dL, and LDL-C > 200mg/dL (253,255). The mechanism for the marked increase in LDL-C levels is unknown. It may be due to a genetic predisposition in certain individuals (Apo E2/E2 genotype or high polygenic risk score for hypercholesterolemia) (256). Therefore, it is important for clinicians to monitor lipid levels in patients electing to follow a very low CHO/high fat diet.    

 

Comparison of Low Fat vs. Low Carbohydrate Weight Loss Diets

 

Numerous randomized studies have compared the effect of low fat vs. low CHO weight loss diets on lipid levels. In a study by Foster et al 154 obese individuals were randomized to a low-fat diet and 153 obese individuals to a low CHO diet (259). In the low CHO diet during the first 12 weeks of treatment participants were instructed to limit CHO intake to 20 grams/day in the form of low–glycemic index vegetables after which the diet was gradually liberalized. In the low-fat diet participants were instructed to limit energy intake with approximately 55% of calories from CHO, 30% from fat, and 15% from protein. Participants were instructed to limit calorie intake, with a focus on decreasing fat intake. After 6 months weight loss was similar in both diet groups. The effect on lipid levels at 6 months is shown in table 17. As one would expect the low CHO was very effective at lowering TG levels and increasing HDL-C levels while the low-fat diet was very effective at lowering LDL-C levels. The large weight loss in this trial may have contributed to the large reduction in lipid levels. A review of a large number of meta-analyses comparing a low CHO diet vs. low fat weight loss diet similarly described that the low CHO diet lowered TG levels and increased HDL-C and LDL-C levels compared to the low-fat diet (244). Note the increase in LDL-C with the low-CHO diet was blunted in patients with diabetes or pre-diabetes (244). Also, the increase in LDL-C levels is likely to be greater in low CHO diets that are enriched in SFA (244).

 

Table 17. Comparison of Low Fat vs. Low Carbohydrate Weight Loss Diet on Lipid Levels

 

Low Fat Diet

Low Carbohydrate Diet

 

Weight

-11.3kg

-12.2kg

NS

TGs

-24mg/dL

-49mg/dL

P<0.001

LDL-C

-9.5mg/dL

0.5mg/dL

P<0.001

HDL-C

1.0mg/dL

6.2mg/dL

P<0.001

 

Comparison of Vegetarian and Omnivore Diet on Lipid Levels

 

Vegetarian diets exclude all animal flesh. A meta-analysis of 19 studies comparing a vegetarian vs. omnivore diet found that consumption of vegetarian diets resulted in a 12.2mg/dL decrease in LDL-C (p < 0.001) and 3.4mg/dL decrease in HDL-C (p < 0.001) and a nonsignificant increase in TG levels (5.8 mg/dL; P = 0.090) compared with consumption of an omnivorous diet (260). Vegan diets, which exclude all animal products, were associated with larger LDL-C reductions than lacto-ovo vegetarian diets. A meta-analysis of 11 clinical trials comparing a vegetarian vs. omnivore diet observed similar results (LDL‐C decreased 13.3mg/dL ; P<0.001; HDL decreased 3.9mg/dL; P<0.001) (261). It is likely that a decrease in dietary SFA and cholesterol and an increase in dietary fiber and phytosterols account for the differences in a vegetarian and omnivore diets.

 

Comparison of 14 Different Diets on Lipid Levels

 

In a network meta-analysis of 121 eligible trials with 21, 942 overweight or obese patients Ge and colleagues compared the effect of 14 different diets on LDL-C and HDL-C levels (236). The diets could be grouped into low CHO diets (Atkins, South Beach, Zone), moderate macronutrients diets (Biggest Loser, DASH, Jenny Craig, Mediterranean, Portfolio, Slimming World, Volumetrics, Weight Watchers), and low-fat diets (Ornish, Rosemary Conley). The effect of these different diets on LDL-C and HDL-C levels are shown in table 18. It should be noted that despite considerable weight loss the effect of these diets on LDL-C and HDL-C levels was very modest except for the LDL-C lowering seen with the Portfolio diet. Unfortunately, a comparison of the effect of these diets on TG levels was not reported.

 

Table 18. Effect of Different Diets in Comparison with Usual Diet

Diet vs. Usual Diet

Decrease in Weight (Kg)

Change in LDL-C (mg/dL)

Change in HDL-C (mg/dL)

Atkins

5.46

+2.75

-3.41

Zone

4.07

+2.89

+0.33

Dash

3.63

-3.93

+1.90

Mediterranean

2.87

-4.59

+0.61

Paleolithic

5.31

-7.27

+2.52

Low Fat

4.87

-1.92

+2.13

Jenny Craig

7.77

-0.21

+2.85

Volumetrics

5.95

-7.13

+0.13

Weight Watchers

3.90

-7.13

+0.88

Rosemary Conley

3.76

-7.15

+2.04

Ornish

3.64

-4.71

+4.87

Portfolio

3.64

-21.29

+3.26

Biggest Loser

2.88

-3.90

+0.01

Slimming World

2.15

N/A

N/A

South Beach

9.86

+0.64

-3.60

Dietary Advice

0.31

+2.01

+1.71

 Summary of results of popular named diets network meta-analysis for outcomes at six months

 

In a study carried out in a single center the Atkins, Zone, Weight Watchers, and Ornish diets were compared and the effect on TG levels was also reported (262). Table 19 shows the results of this study at 2 months, a period at which dietary compliance was still high. The magnitude of weight loss was similar but the decrease in LDL-C that occurs with weight loss was blunted with a diet that was high in fat (Atkins diet). In contrast HDL-C levels increased with a high fat diet, particularly SFA (Atkins diet) and decreased with a very low-fat diet (Ornish diet). The weight loss induced decrease in TG levels was blunted by a high CHO intake (Ornish diet). These observations confirm and extend the results described above.

 

Table 19. Effect of Different Diets on Lipid Levels

 

Weight (kg)

LDL-C (mg/dL)

HDL-C (mg/dL)

TG (mg/dL)

Atkins

-3.6

1.3

3.2

-32

Zone

-3.8

-9.7

1.8

-54

Weight Watchers

-3.5

-12.1

-0.2

-9.2

Ornish

-3.6

-16.5

-3.6

-0.4

 

Summary

 

While diets can significantly affect lipid levels it should be recognized that the effect is typically relatively modest compared to drug therapy. Whether these modest effects on lipid levels can reduce the risk of CVD has not been tested in randomized controlled trials and given the difficulty of carrying out such long-term diet studies is likely not to be attempted. However, diet therapy can be initiated early in life and has the potential to result in long-term decreases in lipid levels. Given that studies have shown that long-term modest reductions in LDL-C levels can have major effects on the risk of CVD (a 10mg/dL life-long decrease in LDL-C due to polymorphisms in ATP citrate lyase, HMGCoA reductase, LDL receptor, PCSK9, and NPC1L1 resulted in a 16%-18% decrease in cardiovascular events (263)) it is likely that a similar long-term decrease induced by dietary changes would also be effective in decreasing CVD. A life-long 70mg/dL decrease in TG levels due to polymorphisms in the lipoprotein lipase gene resulted in a 23% decrease in coronary heart disease suggesting that long-term decreases in TG levels due to dietary changes would also be beneficial (264). Thus, long-term reductions in lipid levels induced by diet therapy may reduce the lifetime risk of developing CVD.

 

CURRENT DIETARY GUIDELINES

 

Most dietary guidelines recommended to the general population to prevent disease are very similar so I will only present the recommendations of two organizations. A brief summary of the Guidelines for Americans 2020-2025 is shown in table 20 and the guidelines from the American College of Cardiology/American Heart Association are shown in table 21.

 

Table 20. Guidelines for Americans 2020-2025

Recommend

Limit

Vegetables of all types—dark green; red and orange; beans, peas, and lentils; starchy; and other vegetables

Added sugars—Less than 10 percent of calories per day

Fruits, especially whole fruit

Saturated fat—Less than 10 percent of calories per day

Grains, at least half of which are whole grain

Sodium—Less than 2,300 milligrams per day

Dairy, including fat-free or low-fat milk, yogurt, and cheese, and/or lactose-free versions and fortified soy beverages and yogurt as alternatives

Alcoholic beverages—Adults can

choose not to drink or to drink in moderation by limiting intake to 2 drinks or less in a day for men and 1 drink or less in a day for women

Protein foods, including lean meats, poultry, and eggs; seafood; beans, peas, and lentils; and nuts, seeds, and soy products

 

Oils, including vegetable oils and oils in food, such as seafood and nuts

 

Full guideline is available at DietaryGuidelines.gov

 

Table 21. ACC/AHA Dietary Recommendations to Reduce Risk of ASCVD (265)

1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish is recommended

2. Replacement of saturated fat with dietary monounsaturated and polyunsaturated fats can be beneficial

3. A diet containing reduced amounts of cholesterol and sodium can be beneficial

4. As a part of a healthy diet, it is reasonable to minimize the intake of processed meats, refined carbohydrates, and sweetened beverages

5. As a part of a healthy diet, the intake of trans fats should be avoided

ASCVD- Atherosclerotic CVD

 

DIETARY RECOMMENDATIONS FOR PATIENTS WITH LIPID DISORDERS

 

Elevated LDL-C

 

The dietary approach to reduce LDL-C levels is to avoid TFA and decrease SFA and cholesterol intake while increasing intake of fiber and phytosterols (266). Additionally, weight loss if appropriate can be helpful in lowering LDL-C levels (266). Certain foods are effective in lowering LDL-C levels such as tree nuts or peanuts, plant protein from soy products, beans, peas, chickpeas, or lentils, and viscous soluble fiber from oats, barley, psyllium, eggplant, okra, apples, oranges, or berries and if possible, can be added to the individual’s diet (236,241). If one combines multiple nutritional changes one can have significant reductions in LDL-C levels (table 22).

 

Table 22. Effect of Multiple LDL-C Lowering Changes on LDL-C Levels

Nutritional Intervention

Estimated LDL-C Decrease

Replace 5% of energy from SFA with MUFA or PUFA

5% to 10%

7.5 grams/day viscous fiber

6% to 9%

2 grams/day plant sterols/stanols

5% to 8%

Replace 30 grams animal protein or CHO with plant protein

3% to 5%

Loss 5% body weight if excess adiposity

3% to 5%

Total Effect

22% to 37%

Table adapted from (267).

 

While diet alone usually does not reduce LDL-C sufficiently it adds to the beneficial effect of cholesterol lowering drugs. In a comparison of LDL-C lowering a low-fat diet alone lowered LDL-C by 5%, a statin alone by 27%, and the combination of low-fat diet plus statin by 32% demonstrating an independent and additive effect of combining diet and lipid lowering medications (268). 

 

Modestly Elevated Triglycerides

 

The dietary approach to reduce TG levels is to reduce CHO intake particularly simple and refined sugars and to avoid or minimize alcohol intake (266). Weight loss if appropriate can be very helpful in lowering TG levels (25,266).

 

Markedly Elevated Triglycerides

 

In patients with marked elevations in TGs due to the Familial Chylomicronemia Syndrome a diet very low in fat is often necessary to prevent episodes of pancreatitis (<10% of calories from fat) (269). In patients with this disorder medium chain TGs may be helpful. In patients with the Multifactorial Chylomicronemia Syndrome who present with markedly elevated TGs (>1000mg/dL) initial dietary treatment should be a very low-fat diet until the TGs decrease. Once the TGs decrease one can initiate the diet described above for individuals with modestly elevated TGs.

 

Elevated Lipoprotein (a)

 

There is no evidence that healthy dietary changes significantly lower Lp(a) levels (62,270) . In fact, it should be noted that reducing SFA intake while decreasing LDL-C levels increases Lp(a) levels (271). In certain patients with high Lp(a) levels one may need to balance the benefits of decreasing LDL-C levels with the risks of increasing Lp(a) levels (271).

 

Effect of Dietary Advice on Lipid and Lipoprotein Levels

 

In a meta-analysis of 44 randomized studies with 18,175 healthy adult participants comparing dietary advise vs. no or minimal advice found that dietary advice reduced total serum cholesterol by 5.9mg/dL (95% CI 2.3 to 9.0) and LDL-C by 6.2mg/dL (95% CI 3.1 to 9.4) with no change in HDL-C and TG levels (272). In a meta-analysis of 7 studies with 1081 participants that compared consultation with a dietician vs. usual care there was no difference in the absolute change in TC, LDL-C, or HDL-C levels but TG levels were decreased by 19.4mg/dL (95%CI -37.8 to -1.8; p=0.03) (273). Similarly, in a meta-analysis of 5 randomized trials in 912 patients with type 2 diabetes found that dietary advice from a dietician vs. usual care resulted in a small decrease in LDL-C (6.6mg/dL) in the group receiving advice from the dietician (274). Finally, as discussed earlier the Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) with increased consumption of vegetables, fruits, and grains. LDL-C levels were reduced by 3.55 mg/dL in the intervention group while levels of HDL-C and TGs were not significantly different in the intervention vs comparison groups. Taken together these studies illustrate that diet therapy under many circumstances has only modest effects on lipid and lipoprotein levels. Of course, there are studies and individual patients where major reductions in lipid levels occur. For example, in a life style modification study including a vegetarian diet by Ornish and colleagues a marked decrease in LDL-C was observed (153mg/dL decreasing to 96mg/dL) (58). One is most likely to see dramatic effects the greater the change in diet (for example going from a typical Western diet to a vegetarian low-fat diet) and the higher the baseline lipid levels. Patient ability to follow the dietary advise is crucial for success.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Ioannidis JPA. The Challenge of Reforming Nutritional Epidemiologic Research. JAMA 2018; 320:969-970
  2. Nissen SE. U.S. Dietary Guidelines: An Evidence-Free Zone. Ann Intern Med 2016; 164:558-559
  3. Lichtenstein AH. Nutrient supplements and cardiovascular disease: a heartbreaking story. J Lipid Res 2009; 50 Suppl:S429-433
  4. Rimm EB, Stampfer MJ, Ascherio A, Giovannucci E, Colditz GA, Willett WC. Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med 1993; 328:1450-1456
  5. Kushi LH, Folsom AR, Prineas RJ, Mink PJ, Wu Y, Bostick RM. Dietary antioxidant vitamins and death from coronary heart disease in postmenopausal women. N Engl J Med 1996; 334:1156-1162
  6. Stampfer MJ, Hennekens CH, Manson JE, Colditz GA, Rosner B, Willett WC. Vitamin E consumption and the risk of coronary disease in women. N Engl J Med 1993; 328:1444-1449
  7. Lee CH, Chan RSM, Wan HYL, Woo YC, Cheung CYY, Fong CHY, Cheung BMY, Lam TH, Janus E, Woo J, Lam KSL. Dietary Intake of Anti-Oxidant Vitamins A, C, and E Is Inversely Associated with Adverse Cardiovascular Outcomes in Chinese-A 22-Years Population-Based Prospective Study. Nutrients 2018; 10
  8. Knekt P, Reunanen A, Jarvinen R, Seppanen R, Heliovaara M, Aromaa A. Antioxidant vitamin intake and coronary mortality in a longitudinal population study. Am J Epidemiol 1994; 139:1180-1189
  9. Lee IM, Cook NR, Gaziano JM, Gordon D, Ridker PM, Manson JE, Hennekens CH, Buring JE. Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA 2005; 294:56-65
  10. de Gaetano G. Low-dose aspirin and vitamin E in people at cardiovascular risk: a randomised trial in general practice. Collaborative Group of the Primary Prevention Project. Lancet 2001; 357:89-95
  11. Sesso HD, Buring JE, Christen WG, Kurth T, Belanger C, MacFadyen J, Bubes V, Manson JE, Glynn RJ, Gaziano JM. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians' Health Study II randomized controlled trial. JAMA 2008; 300:2123-2133
  12. Heart Protection Study Collaborative G. MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:23-33
  13. Rimm EB, Willett WC, Hu FB, Sampson L, Colditz GA, Manson JE, Hennekens C, Stampfer MJ. Folate and vitamin B6 from diet and supplements in relation to risk of coronary heart disease among women. JAMA 1998; 279:359-364
  14. Voutilainen S, Rissanen TH, Virtanen J, Lakka TA, Salonen JT. Low dietary folate intake is associated with an excess incidence of acute coronary events: The Kuopio Ischemic Heart Disease Risk Factor Study. Circulation 2001; 103:2674-2680
  15. Jayedi A, Zargar MS. Intake of vitamin B6, folate, and vitamin B12 and risk of coronary heart disease: a systematic review and dose-response meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2019; 59:2697-2707
  16. Armitage JM, Bowman L, Clarke RJ, Wallendszus K, Bulbulia R, Rahimi K, Haynes R, Parish S, Sleight P, Peto R, Collins R. Effects of homocysteine-lowering with folic acid plus vitamin B12 vs placebo on mortality and major morbidity in myocardial infarction survivors: a randomized trial. JAMA 2010; 303:2486-2494
  17. Albert CM, Cook NR, Gaziano JM, Zaharris E, MacFadyen J, Danielson E, Buring JE, Manson JE. Effect of folic acid and B vitamins on risk of cardiovascular events and total mortality among women at high risk for cardiovascular disease: a randomized trial. JAMA 2008; 299:2027-2036
  18. Lonn E, Yusuf S, Arnold MJ, Sheridan P, Pogue J, Micks M, McQueen MJ, Probstfield J, Fodor G, Held C, Genest J, Jr. Homocysteine lowering with folic acid and B vitamins in vascular disease. N Engl J Med 2006; 354:1567-1577
  19. Marti-Carvajal AJ, Sola I, Lathyris D, Dayer M. Homocysteine-lowering interventions for preventing cardiovascular events. Cochrane Database Syst Rev 2017; 8:CD006612
  20. Zeraatkar D, Han MA, Guyatt GH, Vernooij RWM, El Dib R, Cheung K, Milio K, Zworth M, Bartoszko JJ, Valli C, Rabassa M, Lee Y, Zajac J, Prokop-Dorner A, Lo C, Bala MM, Alonso-Coello P, Hanna SE, Johnston BC. Red and Processed Meat Consumption and Risk for All-Cause Mortality and Cardiometabolic Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171:703-710
  21. Qian F, Riddle MC, Wylie-Rosett J, Hu FB. Red and Processed Meats and Health Risks: How Strong Is the Evidence? Diabetes Care 2020; 43:265-271
  22. Ordovas JM, Lopez-Miranda J, Mata P, Perez-Jimenez F, Lichtenstein AH, Schaefer EJ. Gene-diet interaction in determining plasma lipid response to dietary intervention. Atherosclerosis 1995; 118 Suppl:S11-27
  23. Vazquez-Vidal I, Desmarchelier C, Jones PJH. Nutrigenetics of Blood Cholesterol Concentrations: Towards Personalized Nutrition. Curr Cardiol Rep 2019; 21:38
  24. Flock MR, Green MH, Kris-Etherton PM. Effects of adiposity on plasma lipid response to reductions in dietary saturated fatty acids and cholesterol. Adv Nutr 2011; 2:261-274
  25. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  26. Kris-Etherton PM, Petersen K, Van Horn L. Convincing evidence supports reducing saturated fat to decrease cardiovascular disease risk. BMJ Nutr Prev Health 2018; 1:23-26
  27. Li Y, Hruby A, Bernstein AM, Ley SH, Wang DD, Chiuve SE, Sampson L, Rexrode KM, Rimm EB, Willett WC, Hu FB. Saturated Fats Compared With Unsaturated Fats and Sources of Carbohydrates in Relation to Risk of Coronary Heart Disease: A Prospective Cohort Study. J Am Coll Cardiol 2015; 66:1538-1548
  28. Lichtenstein AH. Dietary Fat and Cardiovascular Disease: Ebb and Flow Over the Last Half Century. Adv Nutr 2019; 10:S332-S339
  29. Jakobsen MU, O'Reilly EJ, Heitmann BL, Pereira MA, Balter K, Fraser GE, Goldbourt U, Hallmans G, Knekt P, Liu S, Pietinen P, Spiegelman D, Stevens J, Virtamo J, Willett WC, Ascherio A. Major types of dietary fat and risk of coronary heart disease: a pooled analysis of 11 cohort studies. Am J Clin Nutr 2009; 89:1425-1432
  30. Farvid MS, Ding M, Pan A, Sun Q, Chiuve SE, Steffen LM, Willett WC, Hu FB. Dietary linoleic acid and risk of coronary heart disease: a systematic review and meta-analysis of prospective cohort studies. Circulation 2014; 130:1568-1578
  31. Zheng Y, Fang Y, Xu X, Ye W, Kang S, Yang K, Cao Y, Xu R, Zheng J, Wang H. Dietary saturated fatty acids increased all-cause and cardiovascular disease mortality in an elderly population: The National Health and Nutrition Examination Survey. Nutr Res 2023; 120:99-114
  32. Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Meta-analysis of prospective cohort studies evaluating the association of saturated fat with cardiovascular disease. Am J Clin Nutr 2010; 91:535-546
  33. Chowdhury R, Warnakula S, Kunutsor S, Crowe F, Ward HA, Johnson L, Franco OH, Butterworth AS, Forouhi NG, Thompson SG, Khaw KT, Mozaffarian D, Danesh J, Di Angelantonio E. Association of dietary, circulating, and supplement fatty acids with coronary risk: a systematic review and meta-analysis. Ann Intern Med 2014; 160:398-406
  34. de Souza RJ, Mente A, Maroleanu A, Cozma AI, Ha V, Kishibe T, Uleryk E, Budylowski P, Schunemann H, Beyene J, Anand SS. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies. BMJ 2015; 351:h3978
  35. Skeaff CM, Miller J. Dietary fat and coronary heart disease: summary of evidence from prospective cohort and randomised controlled trials. Ann Nutr Metab 2009; 55:173-201
  36. Yamagishi K, Iso H, Yatsuya H, Tanabe N, Date C, Kikuchi S, Yamamoto A, Inaba Y, Tamakoshi A. Dietary intake of saturated fatty acids and mortality from cardiovascular disease in Japanese: the Japan Collaborative Cohort Study for Evaluation of Cancer Risk (JACC) Study. Am J Clin Nutr 2010; 92:759-765
  37. Clifton PM, Keogh JB. A systematic review of the effect of dietary saturated and polyunsaturated fat on heart disease. Nutr Metab Cardiovasc Dis 2017; 27:1060-1080
  38. Sacks FM, Lichtenstein AH, Wu JHY, Appel LJ, Creager MA, Kris-Etherton PM, Miller M, Rimm EB, Rudel LL, Robinson JG, Stone NJ, Van Horn LV. Dietary Fats and Cardiovascular Disease: A Presidential Advisory From the American Heart Association. Circulation 2017; 136:e1-e23
  39. Mozaffarian D, Micha R, Wallace S. Effects on coronary heart disease of increasing polyunsaturated fat in place of saturated fat: a systematic review and meta-analysis of randomized controlled trials. PLoS Med 2010; 7:e1000252
  40. de Oliveira Otto MC, Mozaffarian D, Kromhout D, Bertoni AG, Sibley CT, Jacobs DR, Jr., Nettleton JA. Dietary intake of saturated fat by food source and incident cardiovascular disease: the Multi-Ethnic Study of Atherosclerosis. Am J Clin Nutr 2012; 96:397-404
  41. Low-fat diet in myocardial infarction: A controlled trial. Lancet 1965; 2:501-504
  42. Leren P. The Oslo diet-heart study. Eleven-year report. Circulation 1970; 42:935-942
  43. Controlled trial of soya-bean oil in myocardial infarction. Lancet 1968; 2:693-699
  44. Dayton S, Pearce ML, Goldman H, Harnish A, Plotkin D, Shickman M, Winfield M, Zager A, Dixon W. Controlled trial of a diet high in unsaturated fat for prevention of atherosclerotic complications. Lancet 1968; 2:1060-1062
  45. DAYTON S PM, HASHIMOTO S, DIXON WJ, and TOMIYASU U. A Controlled Clinical Trial of a Diet High in Unsaturated Fat in Preventing Complications of Atherosclerosis. Circulation 1969; 40:II-1–II-63
  46. Turpeinen O, Karvonen MJ, Pekkarinen M, Miettinen M, Elosuo R, Paavilainen E. Dietary prevention of coronary heart disease: the Finnish Mental Hospital Study. Int J Epidemiol 1979; 8:99-118
  47. Miettinen M, Turpeinen O, Karvonen MJ, Pekkarinen M, Paavilainen E, Elosuo R. Dietary prevention of coronary heart disease in women: the Finnish mental hospital study. Int J Epidemiol 1983; 12:17-25
  48. Miettinen M, Turpeinen O, Karvonen MJ, Elosuo R, Paavilainen E. Effect of cholesterol-lowering diet on mortality from coronary heart-disease and other causes. A twelve-year clinical trial in men and women. Lancet 1972; 2:835-838
  49. Ramsden CE, Zamora D, Leelarthaepin B, Majchrzak-Hong SF, Faurot KR, Suchindran CM, Ringel A, Davis JM, Hibbeln JR. Use of dietary linoleic acid for secondary prevention of coronary heart disease and death: evaluation of recovered data from the Sydney Diet Heart Study and updated meta-analysis. BMJ 2013; 346:e8707
  50. Burr ML, Fehily AM, Gilbert JF, Rogers S, Holliday RM, Sweetnam PM, Elwood PC, Deadman NM. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: diet and reinfarction trial (DART). Lancet 1989; 2:757-761
  51. Frantz ID, Jr., Dawson EA, Ashman PL, Gatewood LC, Bartsch GE, Kuba K, Brewer ER. Test of effect of lipid lowering by diet on cardiovascular risk. The Minnesota Coronary Survey. Arteriosclerosis 1989; 9:129-135
  52. Howard BV, Van Horn L, Hsia J, Manson JE, Stefanick ML, Wassertheil-Smoller S, Kuller LH, LaCroix AZ, Langer RD, Lasser NL, Lewis CE, Limacher MC, Margolis KL, Mysiw WJ, Ockene JK, Parker LM, Perri MG, Phillips L, Prentice RL, Robbins J, Rossouw JE, Sarto GE, Schatz IJ, Snetselaar LG, Stevens VJ, Tinker LF, Trevisan M, Vitolins MZ, Anderson GL, Assaf AR, Bassford T, Beresford SA, Black HR, Brunner RL, Brzyski RG, Caan B, Chlebowski RT, Gass M, Granek I, Greenland P, Hays J, Heber D, Heiss G, Hendrix SL, Hubbell FA, Johnson KC, Kotchen JM. Low-fat dietary pattern and risk of cardiovascular disease: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 2006; 295:655-666
  53. Wolfe MS, Sawyer JK, Morgan TM, Bullock BC, Rudel LL. Dietary polyunsaturated fat decreases coronary artery atherosclerosis in a pediatric-aged population of African green monkeys. Arterioscler Thromb 1994; 14:587-597
  54. Rudel LL, Parks JS, Sawyer JK. Compared with dietary monounsaturated and saturated fat, polyunsaturated fat protects African green monkeys from coronary artery atherosclerosis. Arterioscler Thromb Vasc Biol 1995; 15:2101-2110
  55. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  56. Silverman MG, Ference BA, Im K, Wiviott SD, Giugliano RP, Grundy SM, Braunwald E, Sabatine MS. Association Between Lowering LDL-C and Cardiovascular Risk Reduction Among Different Therapeutic Interventions: A Systematic Review and Meta-analysis. JAMA 2016; 316:1289-1297
  57. Watts GF, Lewis B, Brunt JN, Lewis ES, Coltart DJ, Smith LD, Mann JI, Swan AV. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St Thomas' Atherosclerosis Regression Study (STARS). Lancet 1992; 339:563-569
  58. Ornish D, Brown SE, Scherwitz LW, Billings JH, Armstrong WT, Ports TA, McLanahan SM, Kirkeeide RL, Brand RJ, Gould KL. Can lifestyle changes reverse coronary heart disease? The Lifestyle Heart Trial. Lancet 1990; 336:129-133
  59. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  60. Mensink RP. Effects of saturated fatty acids on serum lipids and lipoproteins: a systematic review and regression analysis. Geneva, Switzerland: Geneva: World Health Organization; 2016.
  61. Dreon DM, Fernstrom HA, Campos H, Blanche P, Williams PT, Krauss RM. Change in dietary saturated fat intake is correlated with change in mass of large low-density-lipoprotein particles in men. Am J Clin Nutr 1998; 67:828-836
  62. Enkhmaa B, Petersen KS, Kris-Etherton PM, Berglund L. Diet and Lp(a): Does Dietary Change Modify Residual Cardiovascular Risk Conferred by Lp(a)? Nutrients 2020; 12
  63. Mensink RP, Zock PL, Kester AD, Katan MB. Effects of dietary fatty acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr 2003; 77:1146-1155
  64. Wu JHY, Micha R, Mozaffarian D. Dietary fats and cardiometabolic disease: mechanisms and effects on risk factors and outcomes. Nat Rev Cardiol 2019; 16:581-601
  65. Micha R, Mozaffarian D. Saturated fat and cardiometabolic risk factors, coronary heart disease, stroke, and diabetes: a fresh look at the evidence. Lipids 2010; 45:893-905
  66. Flock MR, Kris-Etherton PM. Diverse physiological effects of long-chain saturated fatty acids: implications for cardiovascular disease. Curr Opin Clin Nutr Metab Care 2013; 16:133-140
  67. Fernandez ML, West KL. Mechanisms by which dietary fatty acids modulate plasma lipids. J Nutr 2005; 135:2075-2078
  68. Dietschy JM. Dietary fatty acids and the regulation of plasma low density lipoprotein cholesterol concentrations. J Nutr 1998; 128:444S-448S
  69. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 2002; 109:1125-1131
  70. Mente A, de Koning L, Shannon HS, Anand SS. A systematic review of the evidence supporting a causal link between dietary factors and coronary heart disease. Arch Intern Med 2009; 169:659-669
  71. Schwingshackl L, Hoffmann G. Monounsaturated fatty acids, olive oil and health status: a systematic review and meta-analysis of cohort studies. Lipids Health Dis 2014; 13:154
  72. Zong G, Li Y, Sampson L, Dougherty LW, Willett WC, Wanders AJ, Alssema M, Zock PL, Hu FB, Sun Q. Monounsaturated fats from plant and animal sources in relation to risk of coronary heart disease among US men and women. Am J Clin Nutr 2018; 107:445-453
  73. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med 2013; 368:1279-1290
  74. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  75. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Retraction and Republication: Primary Prevention of Cardiovascular Disease with a Mediterranean Diet. N Engl J Med 2013;368:1279-90. N Engl J Med 2018; 378:2441-2442
  76. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  77. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  78. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785
  79. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  80. Li J, Guasch-Ferre M, Li Y, Hu FB. Dietary intake and biomarkers of linoleic acid and mortality: systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112:150-167
  81. Marklund M, Wu JHY, Imamura F, Del Gobbo LC, Fretts A, de Goede J, Shi P, Tintle N, Wennberg M, Aslibekyan S, Chen TA, de Oliveira Otto MC, Hirakawa Y, Eriksen HH, Kroger J, Laguzzi F, Lankinen M, Murphy RA, Prem K, Samieri C, Virtanen J, Wood AC, Wong K, Yang WS, Zhou X, Baylin A, Boer JMA, Brouwer IA, Campos H, Chaves PHM, Chien KL, de Faire U, Djousse L, Eiriksdottir G, El-Abbadi N, Forouhi NG, Michael Gaziano J, Geleijnse JM, Gigante B, Giles G, Guallar E, Gudnason V, Harris T, Harris WS, Helmer C, Hellenius ML, Hodge A, Hu FB, Jacques PF, Jansson JH, Kalsbeek A, Khaw KT, Koh WP, Laakso M, Leander K, Lin HJ, Lind L, Luben R, Luo J, McKnight B, Mursu J, Ninomiya T, Overvad K, Psaty BM, Rimm E, Schulze MB, Siscovick D, Skjelbo Nielsen M, Smith AV, Steffen BT, Steffen L, Sun Q, Sundstrom J, Tsai MY, Tunstall-Pedoe H, Uusitupa MIJ, van Dam RM, Veenstra J, Monique Verschuren WM, Wareham N, Willett W, Woodward M, Yuan JM, Micha R, Lemaitre RN, Mozaffarian D, Riserus U. Biomarkers of Dietary Omega-6 Fatty Acids and Incident Cardiovascular Disease and Mortality. Circulation 2019; 139:2422-2436
  82. Zhu Y, Bo Y, Liu Y. Dietary total fat, fatty acids intake, and risk of cardiovascular disease: a dose-response meta-analysis of cohort studies. Lipids Health Dis 2019; 18:91
  83. Hooper L, Al-Khudairy L, Abdelhamid AS, Rees K, Brainard JS, Brown TJ, Ajabnoor SM, O'Brien AT, Winstanley LE, Donaldson DH, Song F, Deane KH. Omega-6 fats for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD011094
  84. Abdelhamid AS, Martin N, Bridges C, Brainard JS, Wang X, Brown TJ, Hanson S, Jimoh OF, Ajabnoor SM, Deane KH, Song F, Hooper L. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD012345
  85. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  86. Gardner CD, Kraemer HC. Monounsaturated versus polyunsaturated dietary fat and serum lipids. A meta-analysis. Arterioscler Thromb Vasc Biol 1995; 15:1917-1927
  87. Lichtenstein AH. Dietary trans fatty acids and cardiovascular disease risk: past and present. Curr Atheroscler Rep 2014; 16:433
  88. Mozaffarian D, Katan MB, Ascherio A, Stampfer MJ, Willett WC. Trans fatty acids and cardiovascular disease. N Engl J Med 2006; 354:1601-1613
  89. Nestel P. Trans fatty acids: are its cardiovascular risks fully appreciated? Clin Ther 2014; 36:315-321
  90. Mauger JF, Lichtenstein AH, Ausman LM, Jalbert SM, Jauhiainen M, Ehnholm C, Lamarche B. Effect of different forms of dietary hydrogenated fats on LDL particle size. Am J Clin Nutr 2003; 78:370-375
  91. Brouwer IA, Wanders AJ, Katan MB. Effect of animal and industrial trans fatty acids on HDL and LDL cholesterol levels in humans--a quantitative review. PLoS One 2010; 5:e9434
  92. Bendsen NT, Christensen R, Bartels EM, Astrup A. Consumption of industrial and ruminant trans fatty acids and risk of coronary heart disease: a systematic review and meta-analysis of cohort studies. Eur J Clin Nutr 2011; 65:773-783
  93. Matthan NR, Welty FK, Barrett PH, Harausz C, Dolnikowski GG, Parks JS, Eckel RH, Schaefer EJ, Lichtenstein AH. Dietary hydrogenated fat increases high-density lipoprotein apoA-I catabolism and decreases low-density lipoprotein apoB-100 catabolism in hypercholesterolemic women. Arterioscler Thromb Vasc Biol 2004; 24:1092-1097
  94. van Tol A, Zock PL, van Gent T, Scheek LM, Katan MB. Dietary trans fatty acids increase serum cholesterylester transfer protein activity in man. Atherosclerosis 1995; 115:129-134
  95. Soliman GA. Dietary Cholesterol and the Lack of Evidence in Cardiovascular Disease. Nutrients 2018; 10
  96. Xu Z, McClure ST, Appel LJ. Dietary Cholesterol Intake and Sources among U.S Adults: Results from National Health and Nutrition Examination Surveys (NHANES), 2001(-)2014. Nutrients 2018; 10
  97. Berger S, Raman G, Vishwanathan R, Jacques PF, Johnson EJ. Dietary cholesterol and cardiovascular disease: a systematic review and meta-analysis. Am J Clin Nutr 2015; 102:276-294
  98. Carson JAS, Lichtenstein AH, Anderson CAM, Appel LJ, Kris-Etherton PM, Meyer KA, Petersen K, Polonsky T, Van Horn L. Dietary Cholesterol and Cardiovascular Risk: A Science Advisory From the American Heart Association. Circulation 2020; 141:e39-e53
  99. Hu FB, Stampfer MJ, Manson JE, Rimm E, Colditz GA, Rosner BA, Hennekens CH, Willett WC. Dietary fat intake and the risk of coronary heart disease in women. N Engl J Med 1997; 337:1491-1499
  100. Ascherio A, Rimm EB, Giovannucci EL, Spiegelman D, Stampfer M, Willett WC. Dietary fat and risk of coronary heart disease in men: cohort follow up study in the United States. BMJ 1996; 313:84-90
  101. Alexander DD, Miller PE, Vargas AJ, Weed DL, Cohen SS. Meta-analysis of Egg Consumption and Risk of Coronary Heart Disease and Stroke. J Am Coll Nutr 2016; 35:704-716
  102. Krittanawong C, Narasimhan B, Wang Z, Virk HUH, Farrell AM, Zhang H, Tang WHW. Association Between Egg Consumption and Risk of Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. Am J Med 2021; 134:76-83 e72
  103. Drouin-Chartier JP, Schwab AL, Chen S, Li Y, Sacks FM, Rosner B, Manson JE, Willett WC, Stampfer MJ, Hu FB, Bhupathiraju SN. Egg consumption and risk of type 2 diabetes: findings from 3 large US cohort studies of men and women and a systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112:619-630
  104. Mazidi M, Katsiki N, Mikhailidis DP, Pencina MJ, Banach M. Egg Consumption and Risk of Total and Cause-Specific Mortality: An Individual-Based Cohort Study and Pooling Prospective Studies on Behalf of the Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. J Am Coll Nutr 2019; 38:552-563
  105. Xu L, Lam TH, Jiang CQ, Zhang WS, Zhu F, Jin YL, Woo J, Cheng KK, Thomas GN. Egg consumption and the risk of cardiovascular disease and all-cause mortality: Guangzhou Biobank Cohort Study and meta-analyses. Eur J Nutr 2019; 58:785-796
  106. Zhao B, Gan L, Graubard BI, Mannisto S, Albanes D, Huang J. Associations of Dietary Cholesterol, Serum Cholesterol, and Egg Consumption With Overall and Cause-Specific Mortality: Systematic Review and Updated Meta-Analysis. Circulation 2022; 145:1506-1520
  107. Vincent MJ, Allen B, Palacios OM, Haber LT, Maki KC. Meta-regression analysis of the effects of dietary cholesterol intake on LDL and HDL cholesterol. Am J Clin Nutr 2019; 109:7-16
  108. Herron KL, Lofgren IE, Sharman M, Volek JS, Fernandez ML. High intake of cholesterol results in less atherogenic low-density lipoprotein particles in men and women independent of response classification. Metabolism 2004; 53:823-830
  109. Djousse L, Gaziano JM. Dietary cholesterol and coronary artery disease: a systematic review. Curr Atheroscler Rep 2009; 11:418-422
  110. McNamara DJ. The impact of egg limitations on coronary heart disease risk: do the numbers add up? J Am Coll Nutr 2000; 19:540S-548S
  111. Grundy SM. Does Dietary Cholesterol Matter? Curr Atheroscler Rep 2016; 18:68
  112. Tannock LR, O'Brien KD, Knopp RH, Retzlaff B, Fish B, Wener MH, Kahn SE, Chait A. Cholesterol feeding increases C-reactive protein and serum amyloid A levels in lean insulin-sensitive subjects. Circulation 2005; 111:3058-3062
  113. Jakobsen MU, Dethlefsen C, Joensen AM, Stegger J, Tjonneland A, Schmidt EB, Overvad K. Intake of carbohydrates compared with intake of saturated fatty acids and risk of myocardial infarction: importance of the glycemic index. Am J Clin Nutr 2010; 91:1764-1768
  114. Lambadiari V, Korakas E, Tsimihodimos V. The Impact of Dietary Glycemic Index and Glycemic Load on Postprandial Lipid Kinetics, Dyslipidemia and Cardiovascular Risk. Nutrients 2020; 12
  115. Vega-Lopez S, Venn BJ, Slavin JL. Relevance of the Glycemic Index and Glycemic Load for Body Weight, Diabetes, and Cardiovascular Disease. Nutrients 2018; 10
  116. Jenkins DJA, Dehghan M, Mente A, Bangdiwala SI, Rangarajan S, Srichaikul K, Mohan V, Avezum A, Diaz R, Rosengren A, Lanas F, Lopez-Jaramillo P, Li W, Oguz A, Khatib R, Poirier P, Mohammadifard N, Pepe A, Alhabib KF, Chifamba J, Yusufali AH, Iqbal R, Yeates K, Yusoff K, Ismail N, Teo K, Swaminathan S, Liu X, Zatonska K, Yusuf R, Yusuf S. Glycemic Index, Glycemic Load, and Cardiovascular Disease and Mortality. N Engl J Med 2021; 384:1312-1322
  117. Jenkins DJA, Willett WC, Yusuf S, Hu FB, Glenn AJ, Liu S, Mente A, Miller V, Bangdiwala SI, Gerstein HC, Sieri S, Ferrari P, Patel AV, McCullough ML, Le Marchand L, Freedman ND, Loftfield E, Sinha R, Shu XO, Touvier M, Sawada N, Tsugane S, van den Brandt PA, Shuval K, Khan TA, Paquette M, Sahye-Pudaruth S, Patel D, Siu TFY, Srichaikul K, Kendall CWC, Sievenpiper JL. Association of glycaemic index and glycaemic load with type 2 diabetes, cardiovascular disease, cancer, and all-cause mortality: a meta-analysis of mega cohorts of more than 100 000 participants. Lancet Diabetes Endocrinol 2024; 12:107-118
  118. Yang Q, Zhang Z, Gregg EW, Flanders WD, Merritt R, Hu FB. Added sugar intake and cardiovascular diseases mortality among US adults. JAMA Intern Med 2014; 174:516-524
  119. Narain A, Kwok CS, Mamas MA. Soft drinks and sweetened beverages and the risk of cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Clin Pract 2016; 70:791-805
  120. Ho FK, Gray SR, Welsh P, Petermann-Rocha F, Foster H, Waddell H, Anderson J, Lyall D, Sattar N, Gill JMR, Mathers JC, Pell JP, Celis-Morales C. Associations of fat and carbohydrate intake with cardiovascular disease and mortality: prospective cohort study of UK Biobank participants. BMJ 2020; 368:m688
  121. Malik VS, Li Y, Pan A, De Koning L, Schernhammer E, Willett WC, Hu FB. Long-Term Consumption of Sugar-Sweetened and Artificially Sweetened Beverages and Risk of Mortality in US Adults. Circulation 2019; 139:2113-2125
  122. de Koning L, Malik VS, Kellogg MD, Rimm EB, Willett WC, Hu FB. Sweetened beverage consumption, incident coronary heart disease, and biomarkers of risk in men. Circulation 2012; 125:1735-1741, S1731
  123. Gerber PA, Berneis K. Regulation of low-density lipoprotein subfractions by carbohydrates. Curr Opin Clin Nutr Metab Care 2012; 15:381-385
  124. Aeberli I, Gerber PA, Hochuli M, Kohler S, Haile SR, Gouni-Berthold I, Berthold HK, Spinas GA, Berneis K. Low to moderate sugar-sweetened beverage consumption impairs glucose and lipid metabolism and promotes inflammation in healthy young men: a randomized controlled trial. Am J Clin Nutr 2011; 94:479-485
  125. Fechner E, Smeets E, Schrauwen P, Mensink RP. The Effects of Different Degrees of Carbohydrate Restriction and Carbohydrate Replacement on Cardiometabolic Risk Markers in Humans-A Systematic Review and Meta-Analysis. Nutrients 2020; 12
  126. Mansoor N, Vinknes KJ, Veierod MB, Retterstol K. Effects of low-carbohydrate diets v. low-fat diets on body weight and cardiovascular risk factors: a meta-analysis of randomised controlled trials. Br J Nutr 2016; 115:466-479
  127. Hu T, Mills KT, Yao L, Demanelis K, Eloustaz M, Yancy WS, Jr., Kelly TN, He J, Bazzano LA. Effects of low-carbohydrate diets versus low-fat diets on metabolic risk factors: a meta-analysis of randomized controlled clinical trials. Am J Epidemiol 2012; 176 Suppl 7:S44-54
  128. Goff LM, Cowland DE, Hooper L, Frost GS. Low glycaemic index diets and blood lipids: a systematic review and meta-analysis of randomised controlled trials. Nutr Metab Cardiovasc Dis 2013; 23:1-10
  129. Schwingshackl L, Neuenschwander M, Hoffmann G, Buyken AE, Schlesinger S. Dietary sugars and cardiometabolic risk factors: a network meta-analysis on isocaloric substitution interventions. Am J Clin Nutr 2020; 111:187-196
  130. Te Morenga LA, Howatson AJ, Jones RM, Mann J. Dietary sugars and cardiometabolic risk: systematic review and meta-analyses of randomized controlled trials of the effects on blood pressure and lipids. Am J Clin Nutr 2014; 100:65-79
  131. Fattore E, Botta F, Agostoni C, Bosetti C. Effects of free sugars on blood pressure and lipids: a systematic review and meta-analysis of nutritional isoenergetic intervention trials. Am J Clin Nutr 2017; 105:42-56
  132. Livesey G, Taylor R. Fructose consumption and consequences for glycation, plasma triacylglycerol, and body weight: meta-analyses and meta-regression models of intervention studies. Am J Clin Nutr 2008; 88:1419-1437
  133. Stanhope KL, Bremer AA, Medici V, Nakajima K, Ito Y, Nakano T, Chen G, Fong TH, Lee V, Menorca RI, Keim NL, Havel PJ. Consumption of fructose and high fructose corn syrup increase postprandial triglycerides, LDL-cholesterol, and apolipoprotein-B in young men and women. J Clin Endocrinol Metab 2011; 96:E1596-1605
  134. Kroemer G, Lopez-Otin C, Madeo F, de Cabo R. Carbotoxicity-Noxious Effects of Carbohydrates. Cell 2018; 175:605-614
  135. Softic S, Meyer JG, Wang GX, Gupta MK, Batista TM, Lauritzen H, Fujisaka S, Serra D, Herrero L, Willoughby J, Fitzgerald K, Ilkayeva O, Newgard CB, Gibson BW, Schilling B, Cohen DE, Kahn CR. Dietary Sugars Alter Hepatic Fatty Acid Oxidation via Transcriptional and Post-translational Modifications of Mitochondrial Proteins. Cell Metab 2019; 30:735-753 e734
  136. Naghshi S, Sadeghi O, Willett WC, Esmaillzadeh A. Dietary intake of total, animal, and plant proteins and risk of all cause, cardiovascular, and cancer mortality: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ 2020; 370:m2412
  137. Chen Z, Glisic M, Song M, Aliahmad HA, Zhang X, Moumdjian AC, Gonzalez-Jaramillo V, van der Schaft N, Bramer WM, Ikram MA, Voortman T. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur J Epidemiol 2020; 35:411-429
  138. Budhathoki S, Sawada N, Iwasaki M, Yamaji T, Goto A, Kotemori A, Ishihara J, Takachi R, Charvat H, Mizoue T, Iso H, Tsugane S. Association of Animal and Plant Protein Intake With All-Cause and Cause-Specific Mortality in a Japanese Cohort. JAMA Intern Med 2019; 179:1509-1518
  139. Qi XX, Shen P. Associations of dietary protein intake with all-cause, cardiovascular disease, and cancer mortality: A systematic review and meta-analysis of cohort studies. Nutr Metab Cardiovasc Dis 2020; 30:1094-1105
  140. Schwingshackl L, Hoffmann G. Long-term effects of low-fat diets either low or high in protein on cardiovascular and metabolic risk factors: a systematic review and meta-analysis. Nutr J 2013; 12:48
  141. Wycherley TP, Moran LJ, Clifton PM, Noakes M, Brinkworth GD. Effects of energy-restricted high-protein, low-fat compared with standard-protein, low-fat diets: a meta-analysis of randomized controlled trials. Am J Clin Nutr 2012; 96:1281-1298
  142. Santesso N, Akl EA, Bianchi M, Mente A, Mustafa R, Heels-Ansdell D, Schunemann HJ. Effects of higher- versus lower-protein diets on health outcomes: a systematic review and meta-analysis. Eur J Clin Nutr 2012; 66:780-788
  143. Threapleton DE, Greenwood DC, Evans CE, Cleghorn CL, Nykjaer C, Woodhead C, Cade JE, Gale CP, Burley VJ. Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ 2013; 347:f6879
  144. Pereira MA, O'Reilly E, Augustsson K, Fraser GE, Goldbourt U, Heitmann BL, Hallmans G, Knekt P, Liu S, Pietinen P, Spiegelman D, Stevens J, Virtamo J, Willett WC, Ascherio A. Dietary fiber and risk of coronary heart disease: a pooled analysis of cohort studies. Arch Intern Med 2004; 164:370-376
  145. Kim Y, Je Y. Dietary fibre intake and mortality from cardiovascular disease and all cancers: A meta-analysis of prospective cohort studies. Arch Cardiovasc Dis 2016; 109:39-54
  146. Reynolds A, Mann J, Cummings J, Winter N, Mete E, Te Morenga L. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet 2019; 393:434-445
  147. Liu L, Wang S, Liu J. Fiber consumption and all-cause, cardiovascular, and cancer mortalities: a systematic review and meta-analysis of cohort studies. Mol Nutr Food Res 2015; 59:139-146
  148. Wu Y, Qian Y, Pan Y, Li P, Yang J, Ye X, Xu G. Association between dietary fiber intake and risk of coronary heart disease: A meta-analysis. Clin Nutr 2015; 34:603-611
  149. Loveman E, Colquitt J, Rees K. Cochrane corner: does increasing intake of dietary fibre help prevent cardiovascular disease? Heart 2016; 102:1607-1609
  150. Hartley L, May MD, Loveman E, Colquitt JL, Rees K. Dietary fibre for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2016:CD011472
  151. Hollaender PL, Ross AB, Kristensen M. Whole-grain and blood lipid changes in apparently healthy adults: a systematic review and meta-analysis of randomized controlled studies. Am J Clin Nutr 2015; 102:556-572
  152. Bell LP, Hectorne K, Reynolds H, Balm TK, Hunninghake DB. Cholesterol-lowering effects of psyllium hydrophilic mucilloid. Adjunct therapy to a prudent diet for patients with mild to moderate hypercholesterolemia. JAMA 1989; 261:3419-3423
  153. Anderson JW, Zettwoch N, Feldman T, Tietyen-Clark J, Oeltgen P, Bishop CW. Cholesterol-lowering effects of psyllium hydrophilic mucilloid for hypercholesterolemic men. Arch Intern Med 1988; 148:292-296
  154. Jovanovski E, Yashpal S, Komishon A, Zurbau A, Blanco Mejia S, Ho HVT, Li D, Sievenpiper J, Duvnjak L, Vuksan V. Effect of psyllium (Plantago ovata) fiber on LDL cholesterol and alternative lipid targets, non-HDL cholesterol and apolipoprotein B: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108:922-932
  155. Brown L, Rosner B, Willett WW, Sacks FM. Cholesterol-lowering effects of dietary fiber: a meta-analysis. Am J Clin Nutr 1999; 69:30-42
  156. Ghavami A, Ziaei R, Talebi S, Barghchi H, Nattagh-Eshtivani E, Moradi S, Rahbarinejad P, Mohammadi H, Ghasemi-Tehrani H, Marx W, Askari G. Soluble Fiber Supplementation and Serum Lipid Profile: A Systematic Review and Dose-Response Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:465-474
  157. Cohn JS, Kamili A, Wat E, Chung RW, Tandy S. Reduction in intestinal cholesterol absorption by various food components: mechanisms and implications. Atheroscler Suppl 2010; 11:45-48
  158. Jesch ED, Carr TP. Food Ingredients That Inhibit Cholesterol Absorption. Prev Nutr Food Sci 2017; 22:67-80
  159. Gunness P, Gidley MJ. Mechanisms underlying the cholesterol-lowering properties of soluble dietary fibre polysaccharides. Food Funct 2010; 1:149-155
  160. Piironeen V, Lampi A. Occurrence and levels of phytosterols in foods. New York Marcel Dekker, Inc.
  161. Ras RT, Geleijnse JM, Trautwein EA. LDL-cholesterol-lowering effect of plant sterols and stanols across different dose ranges: a meta-analysis of randomised controlled studies. Br J Nutr 2014; 112:214-219
  162. Demonty I, Ras RT, van der Knaap HC, Meijer L, Zock PL, Geleijnse JM, Trautwein EA. The effect of plant sterols on serum triglyceride concentrations is dependent on baseline concentrations: a pooled analysis of 12 randomised controlled trials. Eur J Nutr 2013; 52:153-160
  163. Calpe-Berdiel L, Escola-Gil JC, Blanco-Vaca F. New insights into the molecular actions of plant sterols and stanols in cholesterol metabolism. Atherosclerosis 2009; 203:18-31
  164. Micha R, Shulkin ML, Penalvo JL, Khatibzadeh S, Singh GM, Rao M, Fahimi S, Powles J, Mozaffarian D. Etiologic effects and optimal intakes of foods and nutrients for risk of cardiovascular diseases and diabetes: Systematic reviews and meta-analyses from the Nutrition and Chronic Diseases Expert Group (NutriCoDE). PLoS One 2017; 12:e0175149
  165. Bechthold A, Boeing H, Schwedhelm C, Hoffmann G, Knuppel S, Iqbal K, De Henauw S, Michels N, Devleesschauwer B, Schlesinger S, Schwingshackl L. Food groups and risk of coronary heart disease, stroke and heart failure: A systematic review and dose-response meta-analysis of prospective studies. Crit Rev Food Sci Nutr 2019; 59:1071-1090
  166. Vernooij RWM, Zeraatkar D, Han MA, El Dib R, Zworth M, Milio K, Sit D, Lee Y, Gomaa H, Valli C, Swierz MJ, Chang Y, Hanna SE, Brauer PM, Sievenpiper J, de Souza R, Alonso-Coello P, Bala MM, Guyatt GH, Johnston BC. Patterns of Red and Processed Meat Consumption and Risk for Cardiometabolic and Cancer Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171:732-741
  167. Fraser GE, Sabate J, Beeson WL, Strahan TM. A possible protective effect of nut consumption on risk of coronary heart disease. The Adventist Health Study. Arch Intern Med 1992; 152:1416-1424
  168. Ros E. Nuts and novel biomarkers of cardiovascular disease. Am J Clin Nutr 2009; 89:1649S-1656S
  169. Ros E. Health benefits of nut consumption. Nutrients 2010; 2:652-682
  170. Sabate J, Haddad E, Tanzman JS, Jambazian P, Rajaram S. Serum lipid response to the graduated enrichment of a Step I diet with almonds: a randomized feeding trial. Am J Clin Nutr 2003; 77:1379-1384
  171. Sabate J, Oda K, Ros E. Nut consumption and blood lipid levels: a pooled analysis of 25 intervention trials. Arch Intern Med 2010; 170:821-827
  172. Sabate J, Wien M. Nuts, blood lipids and cardiovascular disease. Asia Pac J Clin Nutr 2010; 19:131-136
  173. Banel DK, Hu FB. Effects of walnut consumption on blood lipids and other cardiovascular risk factors: a meta-analysis and systematic review. Am J Clin Nutr 2009; 90:56-63
  174. Guasch-Ferre M, Li J, Hu FB, Salas-Salvado J, Tobias DK. Effects of walnut consumption on blood lipids and other cardiovascular risk factors: an updated meta-analysis and systematic review of controlled trials. Am J Clin Nutr 2018; 108:174-187
  175. Lee-Bravatti MA, Wang J, Avendano EE, King L, Johnson EJ, Raman G. Almond Consumption and Risk Factors for Cardiovascular Disease: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Adv Nutr 2019; 10:1076-1088
  176. Hadi A, Asbaghi O, Kazemi M, Haghighian HK, Pantovic A, Ghaedi E, Abolhasani Zadeh F. Consumption of pistachio nuts positively affects lipid profiles: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2023; 63:5358-5371
  177. Houston L, Probst YC, Chandra Singh M, Neale EP. Tree Nut and Peanut Consumption and Risk of Cardiovascular Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:1029-1049
  178. Pan A, Yu D, Demark-Wahnefried W, Franco OH, Lin X. Meta-analysis of the effects of flaxseed interventions on blood lipids. Am J Clin Nutr 2009; 90:288-297
  179. Hui S, Liu K, Lang H, Liu Y, Wang X, Zhu X, Doucette S, Yi L, Mi M. Comparative effects of different whole grains and brans on blood lipid: a network meta-analysis. Eur J Nutr 2019; 58:2779-2787
  180. Marshall S, Petocz P, Duve E, Abbott K, Cassettari T, Blumfield M, Fayet-Moore F. The Effect of Replacing Refined Grains with Whole Grains on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials with GRADE Clinical Recommendation. J Acad Nutr Diet 2020; 120:1859-1883 e1831
  181. Jenkins DJA, Blanco Mejia S, Chiavaroli L, Viguiliouk E, Li SS, Kendall CWC, Vuksan V, Sievenpiper JL. Cumulative Meta-Analysis of the Soy Effect Over Time. J Am Heart Assoc 2019; 8:e012458
  182. Moradi M, Daneshzad E, Azadbakht L. The effects of isolated soy protein, isolated soy isoflavones and soy protein containing isoflavones on serum lipids in postmenopausal women: A systematic review and meta-analysis. Crit Rev Food Sci Nutr 2020; 60:3414-3428
  183. Tokede OA, Onabanjo TA, Yansane A, Gaziano JM, Djousse L. Soya products and serum lipids: a meta-analysis of randomised controlled trials. Br J Nutr 2015; 114:831-843
  184. Anderson JW, Bush HM. Soy protein effects on serum lipoproteins: a quality assessment and meta-analysis of randomized, controlled studies. J Am Coll Nutr 2011; 30:79-91
  185. Anderson JW, Johnstone BM, Cook-Newell ME. Meta-analysis of the effects of soy protein intake on serum lipids. N Engl J Med 1995; 333:276-282
  186. Reynolds K, Chin A, Lees KA, Nguyen A, Bujnowski D, He J. A meta-analysis of the effect of soy protein supplementation on serum lipids. Am J Cardiol 2006; 98:633-640
  187. Zhan S, Ho SC. Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 2005; 81:397-408
  188. Simental-Mendia LE, Gotto AM, Jr., Atkin SL, Banach M, Pirro M, Sahebkar A. Effect of soy isoflavone supplementation on plasma lipoprotein(a) concentrations: A meta-analysis. J Clin Lipidol 2018; 12:16-24
  189. Hunter PM, Hegele RA. Functional foods and dietary supplements for the management of dyslipidaemia. Nat Rev Endocrinol 2017; 13:278-288
  190. Jenkins DJ, Mirrahimi A, Srichaikul K, Berryman CE, Wang L, Carleton A, Abdulnour S, Sievenpiper JL, Kendall CW, Kris-Etherton PM. Soy protein reduces serum cholesterol by both intrinsic and food displacement mechanisms. J Nutr 2010; 140:2302S-2311S
  191. Ried K, Toben C, Fakler P. Effect of garlic on serum lipids: an updated meta-analysis. Nutr Rev 2013; 71:282-299
  192. Zeng T GF, Zhang C,Song F, Zhao X,  Xi K. A meta‐analysis of randomized, double‐blind, placebo‐controlled trials for the effects of garlic on serum lipid profiles. Journal of the Science of Food and Agricultue 2012; 92:1892-1902
  193. Reinhart KM, Talati R, White CM, Coleman CI. The impact of garlic on lipid parameters: a systematic review and meta-analysis. Nutr Res Rev 2009; 22:39-48
  194. Stevinson C, Pittler MH, Ernst E. Garlic for treating hypercholesterolemia. A meta-analysis of randomized clinical trials. Ann Intern Med 2000; 133:420-429
  195. Kwak JS, Kim JY, Paek JE, Lee YJ, Kim HR, Park DS, Kwon O. Garlic powder intake and cardiovascular risk factors: a meta-analysis of randomized controlled clinical trials. Nutr Res Pract 2014; 8:644-654
  196. Schwingshackl L, Missbach B, Hoffmann G. An umbrella review of garlic intake and risk of cardiovascular disease. Phytomedicine 2016; 23:1127-1133
  197. Sun YE, Wang W, Qin J. Anti-hyperlipidemia of garlic by reducing the level of total cholesterol and low-density lipoprotein: A meta-analysis. Medicine (Baltimore) 2018; 97:e0255
  198. Li S, Guo W, Lau W, Zhang H, Zhan Z, Wang X, Wang H. The association of garlic intake and cardiovascular risk factors: A systematic review and meta-analysis. Crit Rev Food Sci Nutr 2023; 63:8013-8031
  199. Shabani E, Sayemiri K, Mohammadpour M. The effect of garlic on lipid profile and glucose parameters in diabetic patients: A systematic review and meta-analysis. Prim Care Diabetes 2019; 13:28-42
  200. Kitamura K, Okada Y, Okada K, Kawaguchi Y, Nagaoka S. Epigallocatechin gallate induces an up-regulation of LDL receptor accompanied by a reduction of PCSK9 via the annexin A2-independent pathway in HepG2 cells. Mol Nutr Food Res 2017; 61
  201. Cui CJ, Jin JL, Guo LN, Sun J, Wu NQ, Guo YL, Liu G, Dong Q, Li JJ. Beneficial impact of epigallocatechingallate on LDL-C through PCSK9/LDLR pathway by blocking HNF1alpha and activating FoxO3a. J Transl Med 2020; 18:195
  202. Koo SI, Noh SK. Green tea as inhibitor of the intestinal absorption of lipids: potential mechanism for its lipid-lowering effect. J Nutr Biochem 2007; 18:179-183
  203. Onakpoya I, Spencer E, Heneghan C, Thompson M. The effect of green tea on blood pressure and lipid profile: a systematic review and meta-analysis of randomized clinical trials. Nutr Metab Cardiovasc Dis 2014; 24:823-836
  204. Zheng XX, Xu YL, Li SH, Liu XX, Hui R, Huang XH. Green tea intake lowers fasting serum total and LDL cholesterol in adults: a meta-analysis of 14 randomized controlled trials. Am J Clin Nutr 2011; 94:601-610
  205. Hartley L, Flowers N, Holmes J, Clarke A, Stranges S, Hooper L, Rees K. Green and black tea for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2013:CD009934
  206. Zhao Y, Asimi S, Wu K, Zheng J, Li D. Black tea consumption and serum cholesterol concentration: Systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2015; 34:612-619
  207. Liu W, Wan C, Huang Y, Li M. Effects of tea consumption on metabolic syndrome: A systematic review and meta-analysis of randomized clinical trials. Phytother Res 2020; 34:2857-2866
  208. Yuan F, Dong H, Fang K, Gong J, Lu F. Effects of green tea on lipid metabolism in overweight or obese people: A meta-analysis of randomized controlled trials. Mol Nutr Food Res 2018; 62
  209. Igho-Osagie E, Cara K, Wang D, Yao Q, Penkert LP, Cassidy A, Ferruzzi M, Jacques PF, Johnson EJ, Chung M, Wallace T. Short-Term Tea Consumption Is Not Associated with a Reduction in Blood Lipids or Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Nutr 2020; 150:3269-3279
  210. Momose Y, Maeda-Yamamoto M, Nabetani H. Systematic review of green tea epigallocatechin gallate in reducing low-density lipoprotein cholesterol levels of humans. Int J Food Sci Nutr 2016; 67:606-613
  211. Araya-Quintanilla F, Gutierrez-Espinoza H, Moyano-Galvez V, Munoz-Yanez MJ, Pavez L, Garcia K. Effectiveness of black tea versus placebo in subjects with hypercholesterolemia: A PRISMA systematic review and meta-analysis. Diabetes Metab Syndr 2019; 13:2250-2258
  212. Wang D, Chen C, Wang Y, Liu J, Lin R. Effect of black tea consumption on blood cholesterol: a meta-analysis of 15 randomized controlled trials. PLoS One 2014; 9:e107711
  213. Kim A, Chiu A, Barone MK, Avino D, Wang F, Coleman CI, Phung OJ. Green tea catechins decrease total and low-density lipoprotein cholesterol: a systematic review and meta-analysis. J Am Diet Assoc 2011; 111:1720-1729
  214. Xu R, Yang K, Li S, Dai M, Chen G. Effect of green tea consumption on blood lipids: a systematic review and meta-analysis of randomized controlled trials. Nutr J 2020; 19:48
  215. O'Keefe JH, Bhatti SK, Patil HR, DiNicolantonio JJ, Lucan SC, Lavie CJ. Effects of habitual coffee consumption on cardiometabolic disease, cardiovascular health, and all-cause mortality. J Am Coll Cardiol 2013; 62:1043-1051
  216. Urgert R, Katan MB. The cholesterol-raising factor from coffee beans. Annu Rev Nutr 1997; 17:305-324
  217. Jee SH, He J, Appel LJ, Whelton PK, Suh I, Klag MJ. Coffee consumption and serum lipids: a meta-analysis of randomized controlled clinical trials. Am J Epidemiol 2001; 153:353-362
  218. Latham LS, Hensen ZK, Minor DS. Chocolate--guilty pleasure or healthy supplement? J Clin Hypertens (Greenwich) 2014; 16:101-106
  219. Hooper L, Kay C, Abdelhamid A, Kroon PA, Cohn JS, Rimm EB, Cassidy A. Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: a systematic review and meta-analysis of randomized trials. Am J Clin Nutr 2012; 95:740-751
  220. Lin X, Zhang I, Li A, Manson JE, Sesso HD, Wang L, Liu S. Cocoa Flavanol Intake and Biomarkers for Cardiometabolic Health: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Nutr 2016; 146:2325-2333
  221. Shrime MG, Bauer SR, McDonald AC, Chowdhury NH, Coltart CE, Ding EL. Flavonoid-rich cocoa consumption affects multiple cardiovascular risk factors in a meta-analysis of short-term studies. J Nutr 2011; 141:1982-1988
  222. Tokede OA, Gaziano JM, Djousse L. Effects of cocoa products/dark chocolate on serum lipids: a meta-analysis. Eur J Clin Nutr 2011; 65:879-886
  223. Rimm EB, Williams P, Fosher K, Criqui M, Stampfer MJ. Moderate alcohol intake and lower risk of coronary heart disease: meta-analysis of effects on lipids and haemostatic factors. BMJ 1999; 319:1523-1528
  224. Brien SE, Ronksley PE, Turner BJ, Mukamal KJ, Ghali WA. Effect of alcohol consumption on biological markers associated with risk of coronary heart disease: systematic review and meta-analysis of interventional studies. BMJ 2011; 342:d636
  225. Spaggiari G, Cignarelli A, Sansone A, Baldi M, Santi D. To beer or not to beer: A meta-analysis of the effects of beer consumption on cardiovascular health. PLoS One 2020; 15:e0233619
  226. Brinton EA. Effects of ethanol intake on lipoproteins. Curr Atheroscler Rep 2012; 14:108-114
  227. Badia RR, Pradhan RV, Ayers CR, Chandra A, Rohatgi A. The Relationship of Alcohol Consumption and HDL Metabolism in the Multiethnic Dallas Heart Study. J Clin Lipidol 2023; 17:124-130
  228. Kovar J, Zemankova K. Moderate alcohol consumption and triglyceridemia. Physiol Res 2015; 64:S371-375
  229. Klop B, do Rego AT, Cabezas MC. Alcohol and plasma triglycerides. Curr Opin Lipidol 2013; 24:321-326
  230. Cesena FH, Coimbra SR, Andrade AC, da Luz PL. The relationship between body mass index and the variation in plasma levels of triglycerides after short-term red wine consumption. J Clin Lipidol 2011; 5:294-298
  231. Ginsberg H, Olefsky J, Farquhar JW, Reaven GM. Moderate ethanol ingestion and plasma triglyceride levels. A study in normal and hypertriglyceridemic persons. Ann Intern Med 1974; 80:143-149
  232. De Oliveira ESER, Foster D, McGee Harper M, Seidman CE, Smith JD, Breslow JL, Brinton EA. Alcohol consumption raises HDL cholesterol levels by increasing the transport rate of apolipoproteins A-I and A-II. Circulation 2000; 102:2347-2352
  233. You M, Arteel GE. Effect of ethanol on lipid metabolism. J Hepatol 2019; 70:237-248
  234. Sozio M, Crabb DW. Alcohol and lipid metabolism. Am J Physiol Endocrinol Metab 2008; 295:E10-16
  235. Rees K, Takeda A, Martin N, Ellis L, Wijesekara D, Vepa A, Das A, Hartley L, Stranges S. Mediterranean-style diet for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2019; 3:CD009825
  236. Ge L, Sadeghirad B, Ball GDC, da Costa BR, Hitchcock CL, Svendrovski A, Kiflen R, Quadri K, Kwon HY, Karamouzian M, Adams-Webber T, Ahmed W, Damanhoury S, Zeraatkar D, Nikolakopoulou A, Tsuyuki RT, Tian J, Yang K, Guyatt GH, Johnston BC. Comparison of dietary macronutrient patterns of 14 popular named dietary programmes for weight and cardiovascular risk factor reduction in adults: systematic review and network meta-analysis of randomised trials. BMJ 2020; 369:m696
  237. Harsha DW, Sacks FM, Obarzanek E, Svetkey LP, Lin PH, Bray GA, Aickin M, Conlin PR, Miller ER, 3rd, Appel LJ. Effect of dietary sodium intake on blood lipids: results from the DASH-sodium trial. Hypertension 2004; 43:393-398
  238. Siervo M, Lara J, Chowdhury S, Ashor A, Oggioni C, Mathers JC. Effects of the Dietary Approach to Stop Hypertension (DASH) diet on cardiovascular risk factors: a systematic review and meta-analysis. Br J Nutr 2015; 113:1-15
  239. Chiavaroli L, Viguiliouk E, Nishi SK, Blanco Mejia S, Rahelic D, Kahleova H, Salas-Salvado J, Kendall CW, Sievenpiper JL. DASH Dietary Pattern and Cardiometabolic Outcomes: An Umbrella Review of Systematic Reviews and Meta-Analyses. Nutrients 2019; 11
  240. Lari A, Sohouli MH, Fatahi S, Cerqueira HS, Santos HO, Pourrajab B, Rezaei M, Saneie S, Rahideh ST. The effects of the Dietary Approaches to Stop Hypertension (DASH) diet on metabolic risk factors in patients with chronic disease: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2021; 31:2766-2778
  241. Chiavaroli L, Nishi SK, Khan TA, Braunstein CR, Glenn AJ, Mejia SB, Rahelic D, Kahleova H, Salas-Salvado J, Jenkins DJA, Kendall CWC, Sievenpiper JL. Portfolio Dietary Pattern and Cardiovascular Disease: A Systematic Review and Meta-analysis of Controlled Trials. Prog Cardiovasc Dis 2018; 61:43-53
  242. Ramezani-Jolfaie N, Mohammadi M, Salehi-Abargouei A. The effect of healthy Nordic diet on cardio-metabolic markers: a systematic review and meta-analysis of randomized controlled clinical trials. Eur J Nutr 2019; 58:2159-2174
  243. Massara P, Zurbau A, Glenn AJ, Chiavaroli L, Khan TA, Viguiliouk E, Mejia SB, Comelli EM, Chen V, Schwab U, Riserus U, Uusitupa M, Aas AM, Hermansen K, Thorsdottir I, Rahelic D, Kahleova H, Salas-Salvado J, Kendall CWC, Sievenpiper JL. Nordic dietary patterns and cardiometabolic outcomes: a systematic review and meta-analysis of prospective cohort studies and randomised controlled trials. Diabetologia 2022; 65:2011-2031
  244. Kirkpatrick CF, Bolick JP, Kris-Etherton PM, Sikand G, Aspry KE, Soffer DE, Willard KE, Maki KC. Review of current evidence and clinical recommendations on the effects of low-carbohydrate and very-low-carbohydrate (including ketogenic) diets for the management of body weight and other cardiometabolic risk factors: A scientific statement from the National Lipid Association Nutrition and Lifestyle Task Force. J Clin Lipidol 2019; 13:689-711 e681
  245. Kwiterovich PO, Jr., Vining EP, Pyzik P, Skolasky R, Jr., Freeman JM. Effect of a high-fat ketogenic diet on plasma levels of lipids, lipoproteins, and apolipoproteins in children. JAMA 2003; 290:912-920
  246. Buga A, Welton GL, Scott KE, Atwell AD, Haley SJ, Esbenshade NJ, Abraham J, Buxton JD, Ault DL, Raabe AS, Noakes TD, Hyde PN, Volek JS, Prins PJ. The Effects of Carbohydrate versus Fat Restriction on Lipid Profiles in Highly Trained, Recreational Distance Runners: A Randomized, Cross-Over Trial. Nutrients 2022; 14
  247. Buren J, Ericsson M, Damasceno NRT, Sjodin A. A Ketogenic Low-Carbohydrate High-Fat Diet Increases LDL Cholesterol in Healthy, Young, Normal-Weight Women: A Randomized Controlled Feeding Trial. Nutrients 2021; 13
  248. Creighton BC, Hyde PN, Maresh CM, Kraemer WJ, Phinney SD, Volek JS. Paradox of hypercholesterolaemia in highly trained, keto-adapted athletes. BMJ Open Sport Exerc Med 2018; 4:e000429
  249. Retterstol K, Svendsen M, Narverud I, Holven KB. Effect of low carbohydrate high fat diet on LDL cholesterol and gene expression in normal-weight, young adults: A randomized controlled study. Atherosclerosis 2018; 279:52-61
  250. Lima PA, de Brito Sampaio LP, Damasceno NR. Ketogenic diet in epileptic children: impact on lipoproteins and oxidative stress. Nutr Neurosci 2015; 18:337-344
  251. Joo M, Moon S, Lee YS, Kim MG. Effects of very low-carbohydrate ketogenic diets on lipid profiles in normal-weight (body mass index < 25 kg/m2) adults: a meta-analysis. Nutr Rev 2023; 81:1393-1401
  252. Castellana M, Conte E, Cignarelli A, Perrini S, Giustina A, Giovanella L, Giorgino F, Trimboli P. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: A systematic review and meta-analysis. Rev Endocr Metab Disord 2020; 21:5-16
  253. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL Cholesterol with a Carbohydrate-Restricted Diet: Evidence for a "Lean Mass Hyper-Responder" Phenotype. Curr Dev Nutr 2022; 6:nzab144
  254. Houttu V, Grefhorst A, Cohn DM, Levels JHM, Roeters van Lennep J, Stroes ESG, Groen AK, Tromp TR. Severe Dyslipidemia Mimicking Familial Hypercholesterolemia Induced by High-Fat, Low-Carbohydrate Diets: A Critical Review. Nutrients 2023; 15
  255. Norwitz NG, Mindrum MR, Giral P, Kontush A, Soto-Mota A, Wood TR, D'Agostino DP, Manubolu VS, Budoff M, Krauss RM. Elevated LDL-cholesterol levels among lean mass hyper-responders on low-carbohydrate ketogenic diets deserve urgent clinical attention and further research. J Clin Lipidol 2022; 16:765-768
  256. Goldberg IJ, Ibrahim N, Bredefeld C, Foo S, Lim V, Gutman D, Huggins LA, Hegele RA. Ketogenic diets, not for everyone. J Clin Lipidol 2021; 15:61-67
  257. Schaffer AE, D'Alessio DA, Guyton JR. Extreme elevations of low-density lipoprotein cholesterol with very low carbohydrate, high fat diets. J Clin Lipidol 2021; 15:525-526
  258. Schmidt T, Harmon DM, Kludtke E, Mickow A, Simha V, Kopecky S. Dramatic elevation of LDL cholesterol from ketogenic-dieting: A Case Series. Am J Prev Cardiol 2023; 14:100495
  259. Foster GD, Wyatt HR, Hill JO, Makris AP, Rosenbaum DL, Brill C, Stein RI, Mohammed BS, Miller B, Rader DJ, Zemel B, Wadden TA, Tenhave T, Newcomb CW, Klein S. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a randomized trial. Ann Intern Med 2010; 153:147-157
  260. Yokoyama Y, Levin SM, Barnard ND. Association between plant-based diets and plasma lipids: a systematic review and meta-analysis. Nutr Rev 2017; 75:683-698
  261. Wang F, Zheng J, Yang B, Jiang J, Fu Y, Li D. Effects of Vegetarian Diets on Blood Lipids: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Am Heart Assoc 2015; 4:e002408
  262. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the Atkins, Ornish, Weight Watchers, and Zone diets for weight loss and heart disease risk reduction: a randomized trial. JAMA 2005; 293:43-53
  263. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  264. Ference BA, Kastelein JJP, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, Laufs U, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Nicholls SJ, Bhatt DL, Sabatine MS, Catapano AL. Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA 2019; 321:364-373
  265. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Munoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 140:e596-e646
  266. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  267. Kirkpatrick CF, Sikand G, Petersen KS, Anderson CAM, Aspry KE, Bolick JP, Kris-Etherton PM, Maki KC. Nutrition interventions for adults with dyslipidemia: A Clinical Perspective from the National Lipid Association. J Clin Lipidol 2023; 17:428-451
  268. Hunninghake DB, Stein EA, Dujovne CA, Harris WS, Feldman EB, Miller VT, Tobert JA, Laskarzewski PM, Quiter E, Held J, et al. The efficacy of intensive dietary therapy alone or combined with lovastatin in outpatients with hypercholesterolemia. N Engl J Med 1993; 328:1213-1219
  269. Subramanian S. Hypertriglyceridemia: Pathophysiology, Role of Genetics, Consequences, and Treatment. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  270. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  271. Law HG, Meyers FJ, Berglund L, Enkhmaa B. Lipoprotein(a) and diet-a challenge for a role of saturated fat in cardiovascular disease risk reduction? Am J Clin Nutr 2023; 118:23-26
  272. Rees K, Dyakova M, Ward K, Thorogood M, Brunner E. Dietary advice for reducing cardiovascular risk. Cochrane Database Syst Rev 2013:CD002128
  273. Ross LJ, Barnes KA, Ball LE, Mitchell LJ, Sladdin I, Lee P, Williams LT. Effectiveness of dietetic consultation for lowering blood lipid levels in the management of cardiovascular disease risk: A systematic review and meta-analysis of randomised controlled trials. Nutr Diet 2019; 76:199-210
  274. Moller G, Andersen HK, Snorgaard O. A systematic review and meta-analysis of nutrition therapy compared with dietary advice in patients with type 2 diabetes. Am J Clin Nutr 2017; 106:1394-1400