Archives

Estrogens, Male Reproduction and Beyond

ABSTRACT

 

In males, estrogens exert pleiotropic effects by acting on several tissue and organs, including the male reproductive system. The action of estrogens is manifest from prenatal life during which the exposure to estrogen excess might influence the development of some structures of the male reproductive tract. Male fertility is under the control of estrogens, especially in rodents. The loss of function of estrogen receptor alpha and/or of the aromatase enzyme leads to infertility in mice. In men, estrogens are able to exert their actions at several levels through the reproductive tract and on several different reproductive cells. However, the regulation of human male reproduction is complex, and the role of estrogens is less clear compared to mice. During fetal and perinatal life, estrogen acts on the central nervous system by modulating the development of some areas within the brain that are committed to controlling male sexual behavior in terms of setting gender identity, sexual orientation development and the evolution of normal adult male sexual behavior. This organizational, central effect of estrogens is of particular significance in other species (especially rodents and rams), but probably less important in men where psychosocial factors become more determining. Other relevant, non-reproductive physiological events, such as bone maturation and mineralization and glucose metabolism, depend on estrogen in men and an increasing body of evidence is disclosing new non-reproductive estrogen function. In this chapter we provide an update of estrogen’s role by reviewing the physiological actions of estrogen on male reproduction and the pathophysiology related to estrogen deficiency and estrogen excess. Phenotypes associated with estrogen deficiency and excess in rodents and in man have shed new light on the mechanisms involved in male reproduction, challenging the perception of the predominant importance of androgens in men. It is now clear that the imbalance between estrogen and androgen in men might affect male reproductive function even in presence of normal circulating androgens. Some uncertainties still remain, especially regarding the impact of abnormal serum estrogen levels on male health, particularly due to the fact that estrogen is not routinely measured in men in clinical practice. Advancements in methods to precisely measure estrogens in men, together with a reduction of their costs, should provide better evidence on this issue and inform clinical practice. In parallel, new basic, genetic, and clinical research is required to improve our knowledge on the role of estrogen in male reproductive function and men’s health in general.

 

INTRODUCTION

 

From an historical perspective, estrogens were identified about 85 years ago and estradiol was identified in 1940, reviewed in (1,2). The first evidence of estrogen production in the male was provided in 1934 by Zondek (3), who documented that male stallions excrete high levels of estrogens in the urine and hypothesized that estrogen production in the male occurs via the intratesticular conversion of androgens into estrogens (1,3,4).

 

In men, the conversion of androgens into estrogens was first demonstrated a few years later, when an increase in urinary estrogens after the administration of exogenous testosterone was recorded in normal men (5) (Figure 1).

 

Figure 1. Milestones in the advancement of research in the area of estrogens in men.
[E2: 17β-estradiol; ER: estrogen receptor; ERKO: Estrogen Receptor Knock out; ARKO: Aromatase Knock out]

A more detailed demonstration of estrogen production in the human male was provided several years later in 1979 by MacDonald et al. who showed that the aromatization of androgens to estrogens can occur also peripherally in several tissues other than in the testes (2,6).

 

Prior to the demonstration of estrogen production in males, the effects of estrogen excess on the development of male reproductive organs had been evident since the 1930s (7). Thus, the concept that male tissues are responsive to estrogens was not new, but it was thought that only a great amount of estrogens was able to induce such changes in males (2).

 

Notwithstanding the large amount of data accumulated in the last eighty years, research in the field of estrogen excess and its role on male reproductive system is still ongoing (8) (Figure 1).

 

The pioneering studies of Zondek and MacDonald opened the way for an appreciation of the physiological roles of estrogens in the male beyond their effects during embryogenesis (2). Several studies, year after year, provided further data on estrogen’s role in men (9-12), since the first pilot studies on estrogens and male reproductive function (13,14).

 

The progressive development of immunohistochemical studies and the subsequent progress in the field of molecular biology highlighted the actions of estrogens in the male [for further details see (4)] and opened the way for the creation of estrogen null mice, a useful animal model to study the physiological role of estrogens in vivo (15) (Figure 1).

 

The detailed characterization of estrogen receptors’ structure and function (15,16) together with the discovery and the characterization of genes involved in estrogens synthesis (17) disclosed the biomolecular mechanisms and related pathways involved in estrogen function and dysfunction. It is now clear that estrogen effects in the male are not confined to reproductive organs but are pleiotropic (18).

 

In addition, the development of male transgenic mice lacking functional estrogen receptors or the aromatase enzyme (responsible for estrogen biosynthesis) further contributed to advancements in this field (15,16). Finally, the discovery of mutations in both the human estrogen receptor alpha (19) and aromatase (20,21) genes contributed to an understanding of estrogen’s role in human male physiology and pathophysiology (11,12,22-25) (Figure 1).

 

Nowadays, notwithstanding this long history of studies, reviewed in (26-28), the role of estrogens in the physiology of the male reproductive tract is still not fully understood. The presence of estrogens in the human testis is well documented (29,30), and there is clear evidence that estrogens exert a wide range of biological effects in both men and women (10,12,18,24,25,31).

 

PHYSIOLOGY

 

Estrogen Biosynthesis in Males

 

The term estrogen refers to any substance, natural or synthetic, able to interact with the estrogen receptor (ER) (32,33). 17β-estradiol (estradiol) is the prevalent endogenous estrogen form in mammals, although many of its metabolites could be detected with several degrees of estrogenic activity (34). In humans, the three major endogenous estrogens are estrone (E1), estradiol (E2), and estriol (E3) (33) (Figure 2). In males, estrogens mainly derive from circulating androgens. The key step in estrogen biosynthesis is the aromatization of the C19 androgens, testosterone and androstenedione, to form estradiol and estrone, respectively (32). This step is under the control of the aromatase enzyme (32,35) (Figure 2).

Figure 2. Biochemical pathway of testosterone conversion into estrogen.

However, a wide number of other endogenous products belongs to the category of estrogenic compounds, such as 27-hydroxycholesterol, dehydroepiandrosterone (DHEA), 7-oxo-DHEA, 7α-hydroxy-DHEA, 16α-hydroxy-DHEA, 7β-hydroxyepiandrosterone, Δ4-androstenedione, Δ5-androstenediol, 3α-androstanediol (3α-Adiol), 3β-androstanediol (3β-Adiol), 2-hydroxyestradiol, 2-hydroxyestrone, 4-hydroxyestradiol, and 4-hydroxyestrone and 16α-hydroxyestrone(33). In particular, dihydrotestosterone (DHT), an androgenic metabolite of testosterone that is synthesized by the enzyme 5 alpha reductase, can be metabolized into 3 β-Adiol, an intermediate metabolite with estrogenic activity (32,36). All these molecules differ in terms of ER affinity (33). Various exogenous substances also show estrogenic activity, such as bisphenol A, metalloestrogens, phytoestrogens (e.g., coumestrol, daidzein, genistein, miroestrol) and mycoestrogens (e.g., zeranol) (37). These exogenous estrogens can influence human physiology via environmental exposure or ingestion, however the real impact in vivo as well as critical thresholds and cumulative amount of exposure remain to be fully elucidated (38).

 

The aromatase enzyme is a P450 mono-oxygenase enzyme complex (17) present in the smooth endoplasmic reticulum, which acts through three consecutive hydroxylation reactions, with the final reaction being the aromatization of the A ring of androgens (17,34) (Figure 2). This enzymatic complex is composed of a ubiquitous and non-specific NADPH-cytochrome P450 reductase, together with the regulated form of cytochrome P450 aromatase (17,29). The latter is highly specific for androgens (39,40). The conversion of androgens into estrogens takes place mainly in the testes, adipose tissue and muscle tissue, even though other male tissues are also involved to a lesser extent (17,34,35) (Figure 2).

 

The P450 aromatase enzyme is encoded by the CYP19A1 gene: a gene of 123 kb of length, which consists of at least 16 exons and is located on the long arm of chromosome 15 in the q21.2 region in humans (9,17,34) (Figure 3). This gene belongs to the cytochrome P450 superfamily, similar to other enzymes involved in steroidogenesis (32).

 

Figure 3. Schematic representation of the human aromatase (CYP19) gene.
[Red bars: first exons associated with upstream alternative, tissue-specific promoters; yellow bars: coding exons; black bar: heme-binding region].

Circulating estrogens are mainly reversibly bound to sex hormone binding globulin (SHBG), a β-globulin, and, to a lesser degree, to albumin (41). The amount of circulating free estradiol depends on several factors, of which the concentrations of albumin and SHBG are the most important (41). Serum free estradiol may be calculated by a complicated formula using total estradiol, SHBG, and albumin levels or may be measured by means of equilibrium dialysis or centrifugal ultrafiltration methodology; both, however, are too time consuming and expensive to be employed in routine clinical practice (41). When calculating free estradiol, the reliability of the value of total serum estradiol should be considered, since assays commonly used for estradiol in clinical laboratories have poor accuracy when measuring the low serum estrogen characteristic in males (42-44).

 

Estrogen Actions in Males

 

Estrogen action is mediated by interaction with specific nuclear estrogen receptors (ERs), which are ligand-inducible transcription factors regulating the expression of target genes after hormone binding (10,34,45). Two subtypes of ERs have been described: estrogen receptor α (ERα) and the more recently discovered estrogen receptor β (ERβ) (34,45). These two ER subtypes show different ligand specificity and transcriptional activity, and mediate the classical, direct, ligand-dependent pathway involving estrogen response elements in the promoters of targets genes and protein-protein interactions with several transcription factors (45). These two different ERs have different transcriptional activity (46). In particular, ERβ shows a weaker transcriptional activity compared to ERα (45). This difference is due to the presence of different ERβ isoforms, which can modulate estrogen signaling using different pathways and lead to different impacts on the regulation of target genes (45,46). In addition, it should be remarked that the co-expression of both ERα and ERβ in the same cell determines a complex cross-talk finally resulting in the antagonistic effect exerted by ERβ on ERα-dependent transcription (45,46). Thus, the presence/absence of ER subtypes together with their cross-talk determines a cell’s ability to respond to different ligands as well as the regulation of transcription of different target genes (45).

 

ERα in humans is encoded by the ESR1 gene located on the long arm of chromosome 6, while the ESR2 gene encodes ERβ and is located on band q22-24 of human chromosome 14 (45,46). The two ER proteins have a high degree of homology at the amino acid level (45) (Figure 4).

Figure 4. Estrogen receptor gene structure showing the 9 exons (lower panel), cDNA domains (indicating exons), and protein structures of both ERα and ERβ (upper panels: colored boxes denote the different functional domains of the protein).

ERs are nuclear receptors in which structurally and functionally distinct domains are recognized. Estrogens bind the COOH-terminal multifunctional ligand-binding domain (LBD), whereas the DNA-binding domain recognizes and binds DNA (45,46). The NH2-terminal domain, the most variable domain, is involved in the transcriptional activation (45). This domain recruits a range of coregulatory protein complexes to the DNA-bound receptor (45). The two ER forms share a high degree of sequence homology except in their NH2-terminal domains. This specificity accounts for different transcriptional effects on different target genes (45,46). The ER genomic pathway begins with the binding of estrogen to ER (45). This interaction induces conformational changes in the ER, allowing receptor dimerization and subsequent nuclear translocation prior to binding to estrogen response elements or to other regulatory sites within target genes (45,46). Thereafter, the availability of several coregulatory proteins influences the transcriptional response to estrogen (45,46).

 

While it is clear that estrogens regulate transcription via nuclear interaction with their receptors, a non-genomic action of estrogens has been also demonstrated, suggesting a different molecular mechanism involved in some estrogen actions (34,45-48). In vitro studies show a very short latency time between the administration of estrogens and the appearance of its biological effects. These actions seem to be mediated by a cell-surface G protein-coupled receptor, known as GPR30, that does not act through a transcriptional mechanism (34,47,48). Rapid effects of estrogens result from the actions of specific receptors localized most often to the plasma membrane; in particular it seems that a monomeric portion of the ERα is translocated from the nucleus to the plasma membrane (47,48).

 

Recently, immunohistochemical analysis of murine tissues reported the presence of GPR30 in the male reproductive tract, including testes, epididymis, vas deferens, seminal vesicles and prostate (49). Furthermore, a rapid response to estradiol suggests that non-genomic estrogen actions are involved also in human spermatozoa (50,51). The different intracellular pathways of estrogen action are summarized in Table 1.

 

Table 1. Characteristics of Estrogen Actions and Related Biomolecular Pathways

Estrogen Actions

Receptors

Mechanism/Pathway

Final effect

Features

 

 

 

Genomic

(Nuclear actions) 

ERα

Transcriptional: nuclear interaction with estrogen-responsive elements 

Modulation of estrogen target gene expression

Slow effects (minutes or hours)

ERβ

Transcriptional: nuclear interaction with estrogen-responsive elements 

Modulation of estrogen target gene expression

Slow effects (minutes or hours)

Non-genomic (cell membranes actions)

Estrogen receptors on cells membrane (GPR30) 

Cells membrane changes

Changes in ionic transport through cell surface

Rapid effects (seconds)

[ERα: estrogen receptor alpha; ERβ: estrogen receptor beta].

 

Aromatase enzyme and ERs are widely expressed in the male reproductive tract both in animals and humans (52,53), implying that estrogen biosynthesis occurs at this site and that both locally produced and circulating estrogens may interact with ERs in an intracrine/paracrine and/or endocrine fashion (34). Today, it is clear that not only testicular somatic cells, but also germ cells constitute a source of estrogens in human (29,54). Thus, the concept of a key role for estrogen in the male reproductive tract is strongly supported by the ability of the male reproductive structures to produce and respond to estrogens (26,52). In men, the aromatase enzyme and ERs are expressed in several tissues including those involved in male reproduction. The distribution and expression of aromatase and ERs described below concerns the male reproductive organs.

 

Aromatase and ERs in the Male Reproductive Tract

 

The distribution of ERs and aromatase in both the developing and adult male reproductive tract of rodents and humans is summarized below.

 

DISTRIBUTION OF ERs AND AROMATASE IN FETAL RODENTS

 

Aromatase and ERs are found at a very early stage of development in the rodent testis, thus suggesting a role for estrogens in influencing testicular development (4,26,55-57).

 

Leydig cells in fetal rodent testis express ERα before the androgen receptor. Moreover, ERα is abundant in the developing efferent ductules, which are the first male reproductive structures to express ERs during fetal development (58-60). Furthermore, the epididymis also expresses ERα in the fetal rodent. By contrast, it is unclear whether ERα is present within the seminiferous tubules of the fetal testis since conflicting results have been reported in literature (26,29,57).

 

ERβ is found early in fetal testis, particularly in gonocytes, Sertoli and Leydig cells, with the gonocytes showing the highest expression between 10-16 days post coitum (61). This suggests a role for estrogens in their maturation. In addition, ERβ is expressed by rat Wolffian ducts, the structures from which the efferent ductules and epididymis arise (26,57). ERα is widely expressed in efferent ductules from fetal life to adulthood, implying a crucial role in male reproduction that has been well documented in adult rodents (27,52,60). On the other hand, ERβ is mainly expressed during fetal life, suggesting a major role in the development of male reproductive structures until birth (26).

 

A recent study suggests that estradiol is also able to increase the production of stem cell factors by fetal human Sertoli cells, finally resulting in the proliferation and growth of spermatogonial stem cells (62). With this in view, estrogen deficiency during fetal life may conditioning the total amount of spermatogonia available in the future for the spermatogenetic maturation.

 

Aromatase is expressed in both Leydig and Sertoli cells in the fetal rodent testis, but not in gonocytes and immature structures of the seminal tract. ER and aromatase distribution in the fetal testes as summarized in Table 2. The presence of both aromatase and ERs in the developing fetal testis implies a possible involvement of estrogens in the process of differentiation and maturation of developing rodent testis just starting from an early stage of embryogenesis, with ERβ possibly playing a greater role than ERα (53,55,56).

 

Both ERα and ERβ are expressed in the fetal penile tissue and estrogens seem to be important for penile growth as well as for the normal differentiation of the terminal part of the urethra (63,64). In particular estrogen takes part together with androgens in the final fusion of the penile urethra (64) and estrogen deficiency due to both ERs disruption and aromatase deficiency may cause hypospadias in rodents (63-65).

 

Table 2. ERs and Aromatase Distribution in the Rodent Fetal Testis and Efferent Ducts

 

ERα

ERβ

Aromatase

Leydig cells

++

++

+

Sertoli cells

-

++

++

Gonocytes

-

+++

-

Efferent Ducts

+

+

-

Penile tissue

++

++

?

The proposed distribution is based on information from various studies including immunohistochemical and mRNA expression studies.

[ERα: estrogen receptor alpha; ERβ: estrogen receptor beta].

 

DISTRIBUTION OF ERs AND AROMATASE IN ADULT RODENT REPRODUCTIVE TRACT     

 

ERα is expressed (both in terms of mRNA and protein) in the Leydig cells of both adult rats and mice (66) but not in Sertoli cells, and is mainly expressed in the proximal (rete testis, efferent ductules, proximal epididymis), rather than in the distal (corpus and cauda of the epididymis, vas deferens) reproductive ducts (26). However, in neonatal and prepubertal rats, estradiol increases the expression of proteins involved in the proliferation and differentiation of Sertoli cells and of proteins involved in the adhesion of germ cells to Sertoli cells (67). Furthermore, ERα has been immunolocalized in ciliated and non-ciliated cell nuclei of the epididymal epithelium (59,68). This peculiar distribution explains several important estrogen actions in the proximal ducts, especially within the efferent ductules that are small and convoluted tubules connecting the rete testis (an anastomosing network of intricate and tenuous tubules located in the hilum of the testis) to the epididymis (60). In the efferent ductules, estrogens promote fluid reabsorption (52,60,69). Finally, the full-length form of ERα has been detected in purified rat germ cells, using a specific antibody directed against the C-terminal region of the protein (70) (Table 3).

 

ERβ is expressed (both in terms of mRNA and protein) in Leydig and Sertoli cells in adult rodents (26,57,60) and in monkey germ cells (71); furthermore, it is expressed also in epithelial and peritubular cells of efferent ducts (59,68). For many years the presence of ERβ in rodent germ cells has been the subject of some debate due to discrepancies in the results of different immunohistochemical studies (72). Immunolocalization of ERβ in differentiated germ cells of adult rodents has been revealed in various studies (61,73). Conversely, no ERβ immunoreactivity was found in rodent germ cells in other studies (74), while mRNA expression seems to decline from fetal life to adulthood in the rat (72). Nevertheless, ERβ seem to be involved in the regulation of gonocyte multiplication, which is under the influence of growth factors and estradiol (16), suggesting a functional role for ERβ at least in immature male germ cells. In addition, several studies have recently identified several pathways involving the ERβ in germ cells confirming both its presence and activity of these cells (75,76).

 

Table 3. ERs and Aromatase Distribution in the Adult Rodent Testis and Efferent Ducts

 

ERα

ERβ

GPR30

Aromatase

Leydig cells

+

+ / -

+

+++

Sertoli cells

-

+

+

+

Germ cells

Spermatogonia

Pachytene Spermatocytes

Round Spermatids

Spermatozoa

+

+

+

+

+

++

+

+

+

+

+

+

+

+

?

++++

+

+

++

+

Efferent ductules

++++

+

?

+

The proposed distribution is based on information from various studies including immunohistochemical and mRNA expression studies.

[ER α: estrogen receptor alpha; ERβ: estrogen receptor beta; GPR30: G protein-coupled receptor].

 

GPR30 is widely expressed (both in terms of mRNA and protein) in rodent testis (77). In particular, this receptor is expressed in rat Leydig cells (78) and Sertoli cells (79,80), in the spermatogonia GC-1 cell line (81), in rat pachytene spermatocytes (70), and in round spermatids (82).

 

Rodent Leydig cells show higher aromatase expression than Sertoli cells (83). Aromatase is also expressed at high levels in germ cells throughout all stages of maturation, with its expression increasing as germ cells mature into spermatids. Aromatase mRNA expression and enzyme activity  are present in both rat and mouse germ cells from the pachytene spermatocyte stage, and during their subsequent maturation into round spermatids (57,60,83) (Table 3). Carreau et al. demonstrated that aromatase activity in germ cells was more than 50% of that of the whole testis (29). This intensive activity suggests that germ cells may be a major source of estrogen in adult rodents (57,60,83) (Table 3). Specifically, when fully developed spermatids are released from the epithelium, aromatase is present in the residual body (the remains of the spermatid cytoplasm that is removed during spermiation) and is subsequently phagocytosed by the Sertoli cell. Aromatase activity also remains detectable in the cytoplasmic droplet attached to the flagellum when sperm passes through the epididymis, suggesting that mature spermatozoa are able to synthesize their own estrogen as they pass through the efferent ducts (29,84). The ability to synthesize estrogen gradually decreases as the droplet slowly moves to the end of the tail during epididymal transit until it is finally lost. The demonstration of aromatase in sperm is important as it suggests that the sperm itself could control the levels of estrogen present in the luminal fluid, and might directly modulate some functions such as the reabsorption of fluid from the efferent ductules (60).

 

DISTRIBUTION OF ERs AND AROMATASE IN THE HUMAN MALE REPRODUCTIVE SYSTEM

 

ERs are present in human testis and reproductive tract (29,60,85,86). In the male fetus both ERβ and aromatase are expressed in Sertoli, Leydig and germ cells from 13 to 24 weeks, whereas ERα expression is absent (86,87). Furthermore, ERβ immunoreactivity in the epididymis suggests a putative role for locally produced estrogens, the actions of which are likely mediated by ERβ in this site. This supports the importance of estrogens for the prenatal development and function of male reproductive structures, which is well documented in literature (87). In particular, estrogens play an important role in the development of the rete testis, efferent ductules, epididymis, and vas deferens (88).

Aromatase and ERβ, but not ERα, continue to be expressed (both in terms of mRNA and protein) during the prepubertal period in men, but their function during infancy remains unclear, especially if the very low  levels of both circulating and locally produced sex steroids in this period of life is taken into account (89).

 

In adult men, ERα is expressed only in Leydig cells, while ERβ has been documented in both Leydig and Sertoli cells and in the efferent ducts (74) (Table 4). The presence of ERs in the human epididymis is still a matter of debate (27), even though ERα has been detected in the nuclei of epithelial cells of the caput of the epididymis (90), and recent data confirms its presence in the epididymis (86). Both ERs (ERα and β) have been identified in isolated immature germ cells (29). Furthermore, they were localized in mature spermatozoa (91) and in ejaculated spermatozoa (92). Luconi et al. first described an estrogen receptor-related protein in the sperm membrane (50,51). This protein is able to bind steroid hormones and may act through a calcium-calmodulin dependent pathway, accounting for a well-documented rapid non-genomic action (50,51). Subsequently, the expression (both in terms of mRNA and protein) of both ERs in human ejaculated spermatozoa (92,93) reinforced the concept that estrogens are able to modulate the spermatogenic process from its onset within the testes through to the final process of sperm maturation after ejaculation (4,29,92,93). The ERα and ERβ localize to different regions in human sperm, with ERα present in the compact zone in the equatorial segment of the upper post-acrosomal region of the sperm head, and ERβ in the mid-piece, at the site of the mitochondria (57). This confirms that each type of receptor probably has a distinct role in sperm physiology and in the process of fertilization (75,94).

 

Table 4. ERs and Aromatase Distribution in the Human Testis and Efferent Ductules

 

ERα

ERβ

GPR30

Aromatase

Leydig cells

+

+ / -

+

+

Sertoli cells

-

+

+

+

Germ cells

Spermatogonia

Pachytene Spermatocytes

Round Spermatids

Spermatozoa

 

-

+

+

+

 

+

+

+

++

 

+

-

-

-

 

ND

+

+

+

Efferent ductules

+

+

?

+

The proposed distribution is based on information from various studies including immunohistochemical and mRNA expression studies.

[ERα: estrogen receptor alpha; ERβ: estrogen receptor beta; GPR30: G protein-coupled receptor].

 

Of particular interest is the demonstration of differential expression in the human testis of wild type ERβ (ERβ1) and of a human variant form of ERβ, the latter arising from alternate splicing (known as ERβcx, or ERβ2), (95,96). ERβ2 expression seems to be associated with prevalent, negative inhibition of ER action by inhibiting ERα–induced transactivation (97); it is highest in spermatogonia and Sertoli cells in adult men, suggesting that these cells may be "protected" from estrogen action (95,96). Wild type ERβ1 was mostly present in pachytene spermatocytes and round spermatids, which have been proposed to be more estrogen sensitive (26), yet ERβ1 was low in less mature germ cells (95). In addition, the discovery of several splice variants of ERβ (including ERβ4) in human testicular cells suggests a specific and more complex estrogen action on spermatogenesis (96).

 

Besides, the cellular distribution of non-genomic GPR30 estrogen receptor in human testicular biopsies was examined (98). Immunohistochemical analysis of testicular sections identified the GPR30 receptor in the cytoplasm of Leydig cells, Sertoli cells and spermatogonia (98). This pattern of localization was further demonstrated by the analysis of GPR30 expression (both in terms of mRNA and protein) in isolated germ cells and in Sertoli cell culture (98). This peculiar distribution suggests that GPR30 may be involved in germ cell differentiation (98). Furthermore, the presence of GPR30 in human spermatozoa has been confirmed at both the mRNA and protein level, with this receptor being localized in the sperm mid-piece (99). The co-expression of the two classic ERs and of the GPR30 receptor in the same area within the spermatozoa (mid-piece and acrosome region) suggests a complex cross-talk among all these receptors able to influence physiological processes and pathological implications, such as tumorigenesis (100).

 

Aromatase expression in the human testis is present in both somatic and germ cells (53,88). Specifically, it is expressed in Leydig and Sertoli cells (101,102), in immature germ cells, from pachytene spermatocytes through elongated spermatids (57,101), and ejaculated sperm cells (103). Locally produced estrogens in sperm are proposed to exert a protective action on sperm DNA by preventing sperm DNA damage (104), thus accounting for estrogen’s potential role as a survival factor during sperm transit through the seminal vesicles (105). Unlike rodents, aromatase expression in human gametes persists during the transit through the genital tracts, since P450 aromatase has been demonstrated in human ejaculated spermatozoa at three different functional levels: mRNA expression, protein production and activity (92). Therefore, as in rodents, human sperm are considered a potential site of estrogen biosynthesis (4,92,101,102,104). The presence of functional aromatase in human spermatozoa allows the conversion of androgens into estrogens as they transit the reproductive tract, providing free estrogens in the seminal fluid able to act on the cells of the reproductive ducts. Thus, human spermatozoa should be considered a mobile endocrine unit (53,54,88,106).

 

In summary, the testes are able to synthesize and respond to estrogens throughout their development (53,88). The localization of ERα, ERβ and aromatase suggests that estrogen action is likely to be important for testicular and efferent ductule function. Differences among various polymorphisms of ER genes may account for different responses to estrogens in term of sperm count and sperm quality (107,108). The role of estrogens in the male reproductive system is clearer in rodents (see below), and the mapping of ERs and aromatase distribution in the human male reproductive system has led to the suggestion that estrogen plays a role in human male reproduction (4,53,55). As a consequence, a new field of research has evolved, aimed at improving our knowledge on estrogen action on male reproduction, and the molecular mechanisms involved in both animal models and in men.

 

ROLE OF ESTROGENS IN MALE REPRODUCTION

 

Estrogens in Animal Male Reproduction: Effects of Estrogen Deficiency

 

Estrogen-deficient knockout mice are useful models to investigate estrogen action in rodents (16,26). At present, four different lines of estrogen receptor-deficient knockout mice have been generated: 1) ERα knockout (α-ERKO) mice with disrupted ERα gene (109-111); 2) ERβ knockout (β-ERKO) mice, with an inactivated Erβ (112), 3) double ERα and ERβ knockout (αβ-ERKO) mice with non-functioning ERα and ERβ (16), and 4) GPR30 knockout mice (113-115). The αERKO, βERKO and αβERKO mice provide very helpful information on the loss of ER function, leading to estrogen resistance. The knockout of the aromatase gene in aromatase knockout (ArKO) mice is an experimental model useful for investigating the congenital lack of both circulating and locally produced estradiol (16,26,116,117). Estrogen-resistant mice (αERKO, βERKO, and αβERKO) have high levels of circulating estrogens with the non-genomic pathway still likely functional. Aromatase-deficient mice have no circulating estradiol however estrogen receptors could be activated by other estrogenic compounds produced outside the aromatase pathway (e.g. 3β-Adiol) or introduced by diet (e.g. phytoestrogens) (26). Furthermore, in 2009, Sinkevicius et al. created transgenic mice with a G525L point mutation in the ligand-binding domain of ERα (ENERKI mice) (118). This allows differentiation of ligand-dependent vs ligand-independent ER actions since these two different pathways could lead to different actions in vivo. The study of fertility of the ENERKI mouse shows that the efferent ductule fluid reabsorption is regulated by ligand-independent actions of ERα, whereas germ cell production and/or viability requires ligand-dependent ERα actions (118). Recently, Yao et al. mapped the Era-binding sites in the efferent ductules of male mice and they found 12105 peaks, of which about 50% were shared by the androgen receptor (119).

 

Recently, the creation of the knockout mice lacking GPR30 estrogen receptor (113,115) allowed an investigation of the reproductive phenotype of mice lacking a functional GPR30, with the results suggesting a minor role of this receptor in male fertility. GPR30 knockout mice did not show abnormalities of endocrine organs, alterations of spermatogenesis and mating behavior, or decreased fertility (114,120). A detailed study of spermatogenesis in this mouse model is, however, still lacking.

 

Studies on transgenic mice lacking ERs or the aromatase enzyme demonstrate that the lack of estrogen action is compatible with life (22,121). Congenital estrogen deficiency in mice leads to an impairment of male reproductive function ranging from normal fertility with a fully male phenotype in βERKO mice, to complete infertility in both αERKO and αβERKO mice. An intermediate pattern exists for the ArKO mice in which spermatogenesis is normal in young mice, but progressively worsens during aging (16,26,60,69,109-112,116,122). Reproductive characteristics of male mouse models are summarized in Table 5.

 

Table 5. Reproductive Phenotype of Male Mouse Models of Estrogen Deficiency

αERKO 

βERKO 

αβERKO 

ArKO 

Infertility

Fully fertile

Similar to αERKO mice

Normal fertility in young mice, infertility with advancing age

Normal FSH
Elevated LH
Elevated testosterone

Elevated estradiol

 

 

--

 

 

--

Normal FSH
Elevated LH
Elevated testosterone
Undetectable estradiol

Germ cell loss and dilated seminiferous tubules 

Normal testicular histology 

Testicular histology similar to αERKO mice 

Histology of the testis is disrupted with advancing age

Impairment of sexual behavior

Normal sexual behavior

Complete suppression of sexual behavior 

Impairment of sexual behavior

The G protein-coupled receptor (GPR30) knockout mice have normal reproductive phenotype.

[ERKO: estrogen receptor knockout mice; α: estrogen receptor alpha; β: estrogen receptor beta; ArKO: Aromatase knockout mice].

 

Male αERKO mice are infertile as the seminiferous epithelium is atrophic and degenerated, and seminiferous tubules and rete testis are dilated (60,69,111), even though the development of male reproductive tract is largely unaffected (16,109,111,123). The disruption of spermatogenesis is progressive as the testicular histology is normal at postnatal day 10, but starts to degenerate at twenty-thirty days of age (69,111). From 40 to 60 days, tubules are markedly dilated with a corresponding significant increase in testicular volume, while the seminiferous epithelium becomes atrophic (16,69,111). A severe impairment in tubule fluid absorption at efferent ducts level is the cause of infertility in αERKO male mice, and this defect is partially mimicked also by the administration of an anti-estrogen drug in wild-type mice (59,60,69). In the male genital tract, the highest concentration of ERα is in the efferent ducts (69) and the estrogen-dependent fluid reabsorption at this site probably results from estrogen interaction with the ERα that seems to regulate the expression of the Na(+)/H(+) exchanger-3 (NHE3) (59,69). This mechanism appears to be the consequence of the ligand-independent ERα activation (118). In fact, the disruption of ERα, or the use of anti-estrogens, results in a decreased expression of NHE3 mRNA, as well as in a decrease of other proteins involved in water reabsorption, such as aquaporin I (124,125). The lack of fluid reabsorption in the efferent ductules of αERKO male mice and the consequent dilatation induces a retroactive progressive swelling of the seminiferous tubules (27,52,60,69,111,126). The seminiferous tubule damage results from the increased fluid pressure and severely impaired spermatogenesis coupled with testicular atrophy as seen at the age of 150 days of age (16,52,60,69). When germ cells from αERKO mice are transplanted in wild type mice, they show normal development (127). Recently, it has been demonstrated that some genes playing important role in the efferent ductules are regulated by Er both independently by estrogens or in combination with androgens where estrogen responsive elements colocalize with androgen responsive elements (119).

 

The αERKO mouse is also characterized by a reduced number, motility, and fertilizing capacity of the sperm levels (Table 5). In addition, αERKO male mice show increased serum luteinizing hormone (LH) and testosterone as well as Leydig cells hyperplasia, together with normal serum follicle-stimulating hormone (FSH) levels (Table 5) (16).

 

The production of both ArKO (122) and βERKO (112) mice added further insights in this field, supporting the idea that estrogen actions on the male reproductive tract are more complex than previously suggested on the basis of the studies performed on αERKO mice (16). In fact, unlike αERKO mice, male ArKO mice are initially fully fertile (122), but fertility decreases with advancing age (Table 5) (26,116,117). Furthermore, βERKO mice are fully fertile and apparently reproductively normal in adulthood (Table 5) (112).

 

The mechanism involved in the development of infertility in ArKO male mice therefore differs from that of the αERKO mice (26). Transgenic mice models suggest that ligand-independent ERα signaling is essential for concentrating epididymal sperm via regulation of efferent ductule fluid reabsorption, while ligand-dependent ERαis involved in germ cell production and/or viability (118). Thus, the lack of estrogen action at the level of the seminiferous epithelium rather than a problem due to impaired fluid reabsorption probably explains infertility in ArKO male mice (26,50). Accordingly, estradiol seems to be necessary for round spermatid survival and estrogen deficiency seems to promote apoptosis before differentiation into elongated spermatid (26,92,105).

 

Studies of αβERKO mice showed a male phenotype very close to that of αERKO mice characterized by infertility and dilated seminiferous tubules (16,26). On the contrary, βERKO male mice were fully fertile (112). These findings lead to the hypothesis that estrogen activity in the male reproductive tract depends on both the type of estrogen receptor involved, and the site of action through the male reproductive tract. Interestingly, results from mice lacking functional ERs or aromatase point to an important role for estrogen in the maintenance of mating behavior in male mice. For this reason, infertility in αERKO, αβERKO and ArKO mice is at least in part due to the reduction of various components of mating behavior from an early age (Table 5) (16,26).

 

The function of the hypothalamo-pituitary-testicular axis is impaired in both αERKO and ArKO male mice, leading to elevated serum LH levels in the presence of normal values of FSH, while, as expected, testosterone is augmented and estrogens are higher than normal or undetectable in αERKO and ArKO mice, respectively (26). Thus, negative effects on male reproduction are the direct result of estrogen deprivation in the reproductive structures or of indirect changes in the regulation of sex steroid secretion.

 

Taken together, all these studies support the concept that a functional ERα, but not ERβ and GPR30, is needed for the development and maintenance of a normal fertility in male mice (15,16,52,55,59,60,69,111,112). Anyhow, it should be remarked that estrogens are also able to self-regulate all these estrogen-related pathways in the male reproductive tract since estrogen receptor expression is regulated by estradiol in rats. In particular ERα positively regulates the expression of both ERα and ERβ while androgen receptor and ERβ negatively regulates ERs expression (128).

 

Estrogens in Human Male Reproduction: Effects of Estrogen Deficiency

 

The demonstration of wide expression of the aromatase enzyme, ERα and ERβ throughout the male reproductive system and within human sperm underlines the role of estrogens in human male reproductive function (4,29,55,106,129). Accordingly, estrogens seem to modulate sperm maturation (50,129), since spermatozoa express ERα and ERβ, and are responsive to estrogens throughout their journey from the testes to the urethra.

 

The characterization of human diseases leading to estrogen deficiency have increased our knowledge about the role of estrogen in male reproductive function as well as on other important physiological functions ranging from longitudinal growth, bone mass acquisition, and metabolic alterations (24,130-132).

 

Data from human subjects with congenital estrogen deficiency have provided conflicting and confusing results. Fertility has been investigated in only one man with estrogen resistance who exhibited a mutation in ERα, rendering him unable to respond to estrogen, thus he could be considered as a human equivalent of the αERKO mouse. However, this man had normal testicular volumes and normal sperm count but with slightly reduced motility (19) (Table 6). At present only one other patient has been diagnosed with estrogen resistance, but a semen analysis was not available for this 18 years-old boy, a possible impairment of fertility being hypothesized on the basis of low inhibin serum levels, reduced testis volume, and cryptorchidism (133). The human reproductive phenotype seems different from that observed in αERKO mice (16,26,52,69,111) since there was no clinical evidence of obstruction of the efferent ductules in the man with estrogen resistance, different to that observed in the rodent model (19). However, no data on the histology of the testis and efferent ductules is available from these two men with estrogen-resistance (19,133,134).

 

The other human model of estrogen deficiency is congenital aromatase deficiency (135). At present, sixteen men with aromatase deficiency have been described (Table 6 and Table 7) (20,136-149). For most of them the genetic diagnosis (143,144) and/or the clinical description (21,150-152), as well as the following clinical studies (153-160)were performed by our research group. These patients showed a variable degree of impaired spermatogenesis (4,11,143,144,161). The hormonal pattern of the patients affected by aromatase deficiency is summarized in Table 6(4,55,135,162). Testicular size in aromatase-deficient men is normal except for three cases having a smaller testes volume (Table 6), while data on testes volume are lacking in some reports (148). Among the eight patients with semen analysis available, six had normal sperm count (139,140,143,146,148,152) and the remaining two had oligospermia (21,22,138) from moderate (138) to severe (21) (Table 6). Anyhow, moderate to severe asthenospermia without teratospermia was also reported independently from the sperm count (21,22,138-140,152) (Table 6). Sperm count was unavailable in the other eight men with aromatase deficiency due to lack of data, diagnosis made at birth, and cases described in prepubertal age (20,136,137,141,143-145,147,149). Moreover, a variable degree of germ cell arrest, ranging from complete depletion of germ cells to arrest at the stage of primary spermatocytes, was described in three aromatase-deficient men who underwent biopsy of the testes (21,22,150,151) (Table 6).

 

Furthermore, a history of cryptorchidism was present in four patients (22,2%) being bilateral in two cases (145,150)and unilateral in the remaining two (139,140,152). These data suggest a possible role of estrogen in testis descent, although this was not seen in the transgenic mouse models. The small number of cases of cryptorchidism among men with aromatase deficiency does not allow any conclusion concerning a possible relationship between estrogen deficiency and the occurrence of abnormalities in testis development and descent. Besides, hypospadias has been reported in one case (145) and preliminary data speak in favor of estrogen role on penile tissue development during fetal growth (63-65), but this aspect needs to be confirmed by more robust studies.

 

In addition, a clinical condition of aromatase deficiency may be also caused by mutation in the cytochrome P450 oxidoreductase (POR) (163), as previously reviewed (162).

 

It should be remarked, however, that a clear cause-effect relationship between infertility and aromatase deficiency was not demonstrated in all these patients (4,135). For this reason, the different degree of fertility impairment found in men with congenital estrogen deficiency does not allow us to establish with certainty whether sperm abnormalities are a consequence of the lack of estrogen action or are an epiphenomenon. Again, this spermatogenetic pattern is different from that observed in ArKO mice (16,26,52,116,117,122).

 

Table 6. Reproductive Phenotypes of Men with Congenital Estrogen Deficiency

 

Estrogen resistance

Aromatase deficiency

Total subjects

2

16

Subjects diagnosed during adulthood

1

11

Age at diagnosis (mean+DS; min-max)

23 years; 18-28

25.8 + 8.6 years; 1-44

REPRODUCTIVE HORMONES

 

 

LH

High

Normal to high

FSH

High

High

Testosterone

Normal

Normal to high

Estradiol

High

Undetectable

EXTERNAL GENITALIA

 

 

Size testis

Normal

Small to normal

Cryptorchidism

Absent

4 cases

Hypospadias

-

1 case

SEMEN ANALYSIS

 

 

Sperm count

Normozoospermia

Oligo to normozoospermia

Viability

Asthenozoospermia

Asthenozoospermia

Testis biopsy

Not performed

Depletion or germ cell arrest at primary spematocyte level

[LH: luteinizing hormone; FSH: follicle-stimulating hormone]

 

The frequency of sperm abnormalities in these patients together with the results from rodent studies suggests a possible role for estrogen in human spermatogenesis, however this requires further elucidation (4,12,24,30). Our knowledge on estrogen’s role in human male reproduction in vivo remains far from complete. The data available in the literature suggests that the action exerted by estrogens on male reproductive organs is more complex than that seen in mice and that estrogen alone does not directly control spermatogenesis to the same extent than in rodents, but are involved in a more complex and evolved network (26,118,164)

 

In addition to human models of congenital estrogen deficiency, other experimental settings have provided information on the role of estrogens on human male fertility.

 

Studies on the association between ER polymorphisms and infertility in men showed that two polymorphisms of ERα (XbaI and PvuII) are associated with azoospermia, severe oligospermia and impaired sperm motility (165-168) and the multiallele (TA)n polymorphism with male infertility (108). However, data available in literature provides conflicting results since Pvull resulted strongly associated with infertility (166), but also a strong protective factor of male fertility (169), depending on the research setting. The RsaI polymorphism of the ERβ has been associated with male infertility in one study (107), but not confirmed in another study (167). The ERβ (RsaI) polymorphism AluI was also associated with sperm motility, while no association with motility was found for the RsaI polymorphism (167). Thus, the association of polymorphisms of estrogen-related genes with both sperm concentration and motility, but not with sperm morphology, further supports a putative role of estrogen in controlling sperm production and quality (170).

 

Furthermore, the investigation of ERs in the nuclear matrix of human spermatozoa showed a reduction of ER levels in the nucleus of idiopathic infertile men compared to normospermic fertile men (171).

 

Interventional research studies show that the administration of aromatase inhibitors to infertile men with documented impaired testosterone-to-estradiol ratio may result in an improvement of their fertility rate, but further evidence is needed to verify their efficacy and safety (see paragraph below ‘Anti-estrogen treatment in men’ for further details) (172,173). These results suggest that such modulation of estrogen metabolism will influence sex hormone balance and the HPT axis while dissecting out direct effects of estrogen on spermatogenesis in vivo is extremely challenging.

 

It seems that exposure to increasing estradiol concentrations might influence glucose metabolism in spermatozoa and that the increase of aromatase activity and estradiol enhances glucose metabolism in capacitated, but not in non-capacitated sperm (93). Recently, intratesticular T and E2 were strongly correlated to aromatase expression in Leydig cells in infertile men; intratesticular T was higher and E2 lower in men with obstructive azoospermia compared to those with nonobstructive azoospermia, suggesting that an imbalance in aromatase expression and thus in the intratesticular T to E2 ratio might play a role in the pathogenesis of male infertility (174). In addition, serum estradiol was directly correlated with motility, sperm count and sperm morphology in male partners of infertile couples enrolled prospectively and low estradiol entered among risk factors for decreased semen quality in multivariate analysis (175).

 

It seems probable that most of estrogen actions operating in mice, such as regulation of sperm motility, sperm capacitation, acrosome reaction, and sperm metabolism also occur in men, but the contribution of estrogens to these processes is quantitatively less important in humans. It seems likely that most of these processes in humans are also regulated by other factors in a complex crosstalk system involving also estrogens. This could also explain why high amounts of estrogens or the exposure to an excess of environmental estrogens (or to xenoestrogens with high estrogenic potency) could negatively impact on male fertility. For these reasons, it is apparently difficult to reconcile existing data about effects of both estrogen deficiency and excess on male reproductive function (13,31,176-178).

 

Regulation of Gonadotropin Feedback

 

The regulation of gonadotropin feedback is an important and well-documented action of estrogen in males. While testosterone has been classically considered the key hormone for the control of gonadotropin feedback in the male, a role for estrogens was recently clarified in studies performed in normal and GnRH-deficient men.  We now know that ERs are expressed both in the hypothalamus and the pituitary. In particular, GnRH neurons express ERβ but not ERα (179,180), thus the inhibitory effects of estrogens on these cells is mediated through other neuromediators (e.g. kisspeptin, neurokinin B) released by other neurons expressing also the ERα (181). ERs, especially ERα are expressed in gonadotropes cells (182).

 

The response of the hypothalamic-pituitary-gonadal axis to androgens is confirmed by the administration of dihydrotestosterone (DHT), which is able to partially decrease LH and FSH with a concomitant reduction in serum testosterone and estradiol (183). However, the discovery of men with congenital estrogen deficiency has provided further evidence for a relationship between estrogens and gonadotropin secretion also in men (22). In fact, serum gonadotropins are high in all adult patients with aromatase deficiency, notwithstanding normal to increased serum testosterone levels (135), thus implying that estrogens are also important for the regulation of circulating gonadotropins levels in men.

 

The effects of estrogens on gonadotropin secretion have been investigated in GnRH-deficient men whose gonadotropin secretion was normalized by pulsatile GnRH administration. Moreover, in order to determine the precise role of sex steroids on the hypothalamo-pituitary-testicular axis, several studies characterized by the administration of testosterone, testosterone plus testolactone (an aromatase inhibitor), or estradiol have been performed (184,185). Testosterone alone induced a significant decrease in mean basal LH and FSH levels as well as of LH pulse amplitude, demonstrating a direct suppressive effect on the pituitary of testosterone and its metabolites. In general, mean LH levels and LH pulse frequency are suppressed to a greater extent in normal control subjects under testosterone administration, suggesting the involvement of a hypothalamic site of action of testosterone (or its metabolites) in suppressing GnRH secretion. In order to discriminate the impact of testosterone from its aromatized products, both groups of subjects were treated with testosterone plus testolactone. The addition of this aromatase inhibitor completely inhibited the testosterone effect on gonadotropin secretion both in normal and GnRH-deficient men, thus leading to a significant increase in mean LH levels in both groups. The latter was greater in normal men who received testolactone alone than in normal men who received testosterone plus testolactone, thus confirming a direct effect of androgens on gonadotropin secretion in normal men. On the basis of the results of these studies, it is clear that the aromatization of testosterone to estradiol is, at least in part, required for normal gonadotropin feedback at the pituitary level (185). In fact, when the same experimental model was applied using estradiol administration instead of testolactone, mean LH and FSH levels as well as LH pulse amplitude decreased significantly during the treatment (184). These studies have demonstrated an important direct inhibitory effect of estradiol on gonadotropin secretion in both GnRH-deficient and normal men (184,185) and support the concept that, at least in part, the inhibitory effect on gonadotropin secretion is mediated by the conversion of testosterone to estradiol (4,186). Accordingly, the administration of the aromatase inhibitor letrozole to healthy adult males is able to suppress aromatase activity and serum estradiol levels leading to an increase of gonadotropins (187). Only the restoration of normal circulating estrogens, by means of transdermal estrogen administration, normalized gonadotropin secretion in this setting (187). In contrast, it seems that the 5α-reduction of testosterone to DHT does not play a very important role in pituitary secretion of gonadotropins (188); DHT, in fact, slightly decreases LH and FSH only after long-term administration (183).

 

All these studies suggest possible estrogen action at the level of hypothalamus. In order to clarify the role of estrogen on the feedback regulation of gonadotropin secretion at hypothalamic level, Hayes et al. (189) conducted a study involving men affected by idiopathic hypogonadotropic hypogonadism (IHH), whose gonadotropin secretion was normalized by long-term pulsatile GnRH therapy, followed by treatment with the aromatase inhibitor anastrozole. They observed that the inhibition of estradiol synthesis led to an increase in mean gonadotropin levels that was greater in normal subjects than in IIH men, suggesting a hypothalamic involvement. The rise in mean LH concentrations in normal subjects due to anastrozole was due to increased LH pulse frequency and amplitude. The authors concluded that estrogen acts at the hypothalamic level by decreasing GnRH pulse frequency and pituitary responsiveness to GnRH (189). Subsequently, the same group (190) demonstrated that the administration of estradiol in normal subjects, whose endogenous testosterone and estradiol synthesis was inhibited through the use of ketoconazole, reduced mean LH levels by lowering LH pulse frequency, but not amplitude. These authors went on to report that the sex steroid component to FSH negative feedback was not androgenic but rather was mediated by estradiol effects on the frequency of GnRH stimulation (190,191).

 

For many years another important unresolved issue has been the relative role of circulating vs. locally produced estrogens in the control of gonadotropin secretion. Now we know that the effects of circulating estrogen are more relevant than that of locally produced, at both the hypothalamic and pituitary level (187) (Figure 5). Accordingly, the administration of both the aromatase inhibitor letrozole and estradiol at different dosages showed that serum testosterone and gonadotropins were inversely related to circulating estradiol, depending on the dose of exogenous estradiol (187). The serum estradiol required to obtain the same levels of gonadotropins were not different compared to that at baseline, suggesting that aromatase inhibition and the blockade of locally produced estrogens are less important than previously thought (187). In the same year, our group reached the same conclusions using a different approach. In men with aromatase deficiency, we demonstrated that circulating rather than locally produced estrogens are the main inhibitors of LH secretion (157). This implies that the role of locally produced estradiol on gonadotropin feedback at hypothalamic and pituitary levels is relatively modest in vivo (Figure 5). However, the role of locally produced estrogens has been poorly investigated since evaluating the effects of locally produced estrogens in vivo is challenging (186).

 

Data available in the literature demonstrate that (i) circulating estrogens are involved in gonadotropin suppression both at pituitary (187) and hypothalamic level (157,190), and (ii) estrogen effects on hypothalamus are independent from central aromatization, but requires adequate amounts of circulating estrogens in normal healthy men (187), in men with IHH (190,191), and in men with aromatase deficiency (157).

 

The effects of estrogen on gonadotropin secretion at the pituitary level operate from early- to mid-puberty (186,192,193) into old age in men (194). The administration of an aromatase inhibitor (anastrozole 1 mg daily for 10 weeks) to young men aged 15-22 years (192) resulted in a 50% decrease in serum estradiol concentrations, an increase in testosterone concentrations and an increase in both LH and FSH values during the study protocol. These hormonal parameters were restored after the discontinuation of anastrozole treatment (192). In addition, the administration of letrozole increased serum LH levels, LH pulse frequency and amplitude and the response of LH to GnRH administration in boys during early and mid-pubertal phases, confirming that estrogens act at the pituitary level during early phases of puberty (193), the role of estrogens in infancy and at the beginning of puberty remaining less known (186). The same mechanism continues to operate during adulthood and early senescence (195), as shown in fifteen eugonadal men, aged 65 years treated with 2 mg anastrozole for 9 weeks, in which serum FSH and LH levels increased significantly, in spite of an increase in serum testosterone levels (194). Similar results were replicated by using letrozole in older men (173). For these reasons, the use of aromatase inhibitors as blockers of the negative feedback on gonadotropin has been tested as a possible strategy useful for the treatment of late-onset male hypogonadism (196). The rationale was that increasing endogenous serum testosterone through the inhibition of the rate of conversion of testosterone into estradiol led to the consequent LH and FSH increase (196). After the first encouraging results (197-199), this kind of treatment seems to be not effective, especially on large-scale clinical trials and for long periods of time (195,196,200,201).

 

Figure 5. Sex steroid control of gonadotropin secretion after recent advances: estrogens, but not androgens, are the main regulator of gonadotropins and the action of circulating estradiol prevails with respect to that of locally produced estradiol.
[T: testosterone; DHT: dihydrotestosterone; E2: estradiol; GnRH: gonadotropin releasing hormone; LH: luteinizing hormone; FSH: follicle-stimulating hormone]

Previous data suggest that estradiol may modulate GnRH receptor number and function at hypothalamic-pituitary level (202), since ERs were detected in GnRH secreting neurons (203). Moreover, both genomic and non-genomic estrogen actions seems to be involved in the regulation of the gonadotropin feedback in males (203,204), although the precise mechanism remains unclear (205). Nevertheless, it is now well established that androgens need to be converted to estrogens in order to ensure the integrity of the gonadotropin feedback mechanism in men, testosterone itself having a lesser role than previously thought (Figure 5), and circulating estrogen, rather than locally produced estrogen, having a major role at the hypothalamic pituitary level (157,187,191).

 

In a complementary way, our knowledge on the role of estrogens in gonadotropin feedback has been enhanced through studies of men with congenital estrogen deficiency. The description of a man lacking a functional ERα (19)revealed a remarkable hormonal pattern consisting of normal serum testosterone, high estradiol and estrone levels, but increased serum FSH and LH concentrations; the serum testosterone remained in the normal range because of increased aromatization of testosterone to estradiol (Table 6). Other important information about estrogen’s role in the human male came from the discovery of naturally occurring mutations in the aromatase gene. To date, of the sixteen different cases of human male aromatase deficiency that have been described, all were discovered to be aromatase-deficient as adults, except one who was diagnosed as a child (137,141) and another one who was diagnosed at 15 months of age (145) (Table 6). Eight out of fourteen adult patients with aromatase deficiency had increased basal FSH concentrations (20,21,135,138-140,144,150-152,154), two had serum FSH in the upper normal range (143,144), and the remaining four had normal FSH (144,146-148). The subject diagnosed during childhood had normal FSH in infancy (137) and high to normal FSH levels at puberty (141). The unique patient diagnosed early at 15 months had normal serum T, LH, and FSH (145). LH was normal in all aromatase adult patients (138-140,144,146,147,150-152), except for one subject with elevated serum LH (20,136) and two subjects with high to normal LH levels (21,143,154)(Table 6). Serum testosterone concentrations were generally normal or high-to-normal except for the first case described with elevated serum levels (20,136), and two other aromatase-deficient men with testosterone slightly above the normal range (138,139). Conversely, another man with aromatase deficiency presented with low to normal serum testosterone levels (150,156). In all sixteen patients estradiol concentrations were undetectable (20,136-149).  (Table 6). The detection of increased gonadotropin levels despite normal-to-increased serum testosterone levels, in these men, further highlights the key role for estrogen in regulating circulating gonadotropins in men (155,157), In normal men with pharmacologically induced sex steroid deprivation, estradiol but not testosterone, was able to restore normal FSH serum levels (191). Due to the concomitant impairment of the patient's spermatogenesis, complete normalization of serum FSH was not achieved in all aromatase-deficient men during estradiol treatment, even in the presence of physiological levels of circulating estradiol (135), only supraphysiological levels of estrogens were able to normalize FSH (21,135,154,155).

 

A detailed study of the effects of different doses of transdermal estradiol on pituitary function in two men with congenital aromatase deficiency demonstrated that estrogens might control not only basal secretion of gonadotropins but also their responsiveness to GnRH administration (138,155,157). In these studies, estrogen administration to three male patients with aromatase deficiency caused a decrease in both basal and GnRH-stimulated LH, FSH and α-subunit secretion with a dose-dependent response to GnRH administration (138,155,157). In 2006, Rochira et al. (157), demonstrated that estrogen’s effects on LH secretion are exerted both at pituitary and hypothalamic level, as shown by the decrease of basal and GnRH-stimulated secretion of LH and the LH pulse amplitude, and the reduction of the frequency of LH pulses respectively, during estrogen treatment to normalize estradiol serum levels in two aromatase-deficient men. In normal physiology, these data provide evidence that the negative feedback effects of circulating estrogens is more important than estrogen locally produced at the hypothalamic level (157). As previously explained, these data confirm data from healthy men (186).

 

Notwithstanding recent advances in the study of estrogen’s role in males, some difficulties remain when data from men with congenital estrogen deficiency are interpreted, particularly if phenotype heterogeneity is considered (161,186). No abnormalities were found in either gonadotropin secretion or in testis position and size in the patient with congenital aromatase deficiency diagnosed in childhood (137), unlike female newborns (206). For these reasons, the role of estrogens in the hypothalamo-pituitary-testicular axis should become relevant in a later stage of life than infancy in men. Furthermore, the smaller than expected increase in FSH levels (given the prevailing serum testosterone levels and impaired spermatogenesis) in two estrogen-deficient men (157), suggests a possible role of estrogens in priming and maturation of hypothalamus-pituitary-gonadal axis in men (155,156). Thus, the control of gonadotropin feedback exerted by sex steroids during early infancy and childhood remains a matter of debate in the human male (186).

 

In conclusion, estrogens are the main sex steroids involved in the control of gonadotropin secretion in men, testosterone having a minor but determinant role as demonstrated by evidence coming from complete androgen insensitivity (CAIS) syndrome in which serum LH is above normal as a consequence of androgen resistance notwithstanding elevated circulating estradiol (207).

 

Estrogens and Prostate

 

Androgens regulate prostate gland growth and differentiation, particularly during its development. Estrogens also act on prostate growth and differentiation through both ERα, and ERβ (208,209). In rodents, the prostate is sensitive to estrogen exposure during development (210).

 

Studies on animals have helped to better understand estrogen’s role in prostate growth. Studies in mice overexpressing aromatase (AROM+) demonstrated that prostate lobes are significantly reduced as a consequence of estrogen excess (211). On the other hand, aromatase-deficient mice presented a hyperplastic prostate gland probably due to the excess of circulating androgens (212) and consistent with hyperplasia of the epithelial, interstitial and luminal compartments (210). Furthermore, McPherson et al., using tissue recombination and an ERβ-specific agonist, demonstrated that ERβ activation results in an anti-proliferative response not influenced by systemic androgen levels, or activation of ERα (212). Moreover, studies on ArKO mice demonstrated that the administration of an ERβ-specific agonist reverted the existing hyperplastic epithelial pathology (212).

 

In terms of prostate carcinogenesis, it is generally assumed that androgenic hormones play a major role in tumor development, since the prostate gland is an androgen-dependent tissue, as is prostate cancer (213). However, considering the fact that testosterone can be converted to estradiol, and that ERs are present in the prostate epithelium (214), theoretically estrogen might also be involved in the induction of prostate cancer. Some polymorphisms (rs2470152, rs10459592, and rs4775936) of the CYP19A1 aromatase gene were associated with an increased risk of prostate cancer (215,216). Besides, patients with prostate cancer who are carriers of the rs4775936 polymorphism of the CYP19A1 aromatase gene show a significantly shorter time of cancer-specific survival compared to patients who do not carry this polymorphism (215). In line with this Bosland et al. found that combined treatment of rats with estradiol and testosterone lead to an increased incidence of prostate cancer from 35-40% with androgen alone to 90-100% (217). The estrogen pathways that may be involved at the molecular level in the process of prostate carcinogenesis are very complex (209). Several studies demonstrate that both ERα and β are involved in the transduction of estrogen signaling in prostate cancer such as cell proliferation pathways (209). Furthermore, ERβ seems mainly involved in pro-apoptotic pathways (e.g. FOXO3 and p-53), while ERα is involved in chronic inflammation, and the two ERs seem to act differently on oncogenes playing suppressive (ERβ) and oncogenic (ERα) roles (209). Proliferation of prostatic cells seems to be promoted by the activation of the ER while ER and GPER seems to exert an antiproliferative action (218,219). The different effects of each ER on the proliferation of prostate cells may accounts for the contrasting results (proliferative/antiproliferative) available in literature, depending on the prevailing activated pathway. However, estrogens also display a biphasic effects in vitro on prostate cells growth, which is enhanced by low estradiol and inhibited by high dose of estradiol (220). Probably, different pathways are activated in presence of estrogen excess, thus leading to a shift in the final effect on cell growth (218,220). At present studies on estrogen signaling in prostate cancer tissue are also providing promising results in term of the utilization of this signature as biomarker useful to tailor hormonal treatment (218).

 

Prostate was normal in aromatase-deficient men and did not change in volume during estrogen replacement therapy (Carani & Rochira; data not published data). Besides, the administration of selective inhibitors of aromatase are helpful for the evaluation of estrogen in vivo effects on prostate. Recently, the combined therapy with transdermal testosterone and the aromatase inhibitor anastrozole in older men with low or low-to-normal serum testosterone (< 350 ng/dL) prevented the increase of prostate volume, but not that of prostate-specific antigen seen in patients treated with testosterone alone (221). Similarly, high serum estradiol resulted directly related to prostate volume in 239 Chinese men with benign prostatic hyperplasia (222) even though these data are limited by the poor accuracy of estradiol measured by immunometric assay.

 

This is in line with the above-mentioned experimental results suggesting an active role of estrogens in prostate cell proliferation in prostate carcinogenesis. Traditionally, exogenous estrogens have been used for the treatment of prostate cancer since the 1940s thanks to their potent inhibitory effect on the HPT axis resulting in the suppression of circulating testosterone (223). However, diethylstilbestrol (DES) used in the past for prostate cancer was strongly associated with thromboembolic side effects(223). Recently, the use of exogenous estrogens for the therapy of prostate cancer is being reconsidered (224). Transdermal estradiol (patch) seems to be effective in inhibiting gonadotropins and in reducing serum testosterone in men with prostate cancer without increasing cardiovascular events (224).In the near future, if estrogen’s role in the prostate will be further elucidated, new treatment strategies will become available for benign prostate hypertrophy and cancer, especially in men with concomitant hypogonadism (225).

 

Estrogens and Male Sexual Behavior

 

Sex steroids act on several aspects of male sexual behavior (226). Sex steroids, mainly testosterone, modulate adult male sexual behavior in mammals (227). In men, sexual behavior is more complex than in other species since it results also from cognitive processes, cultural environment, and an individual system of beliefs (226,228). Thus, sexual behavior does not depend only on hormonal and genetic prerequisites in men (226,228). Traditionally, it was thought that only testosterone, the male hormone, is responsible for the control of male sexual behavior (229). In the last two decades, the possibility that estrogens may be involved in the control of male sexual behavior has received more attention, and an impact of estradiol on male sexuality has become evident (199).

 

Testosterone is mainly involved in the control of sexual desire and sexual drive and in the facilitation and maintenance of a normal sexual genital response (226). Erections, especially nocturnal erections, are also under the control of androgens (230,231). The role of estrogen on male sexual behavior has been poorly investigated and knowledge derives mainly from studies performed on animals or from rare models of human estrogen deficiency. The increasing interest on the treatment of transgender people (232) and on the cross actions of male and female hormones on both sexual behavior (233) and other physiological functions (234) probably have contributed to a better focus on this area of research. In recent years, however, several in vivo experimental settings have addressed this issue. As a result, nowadays all studies on steroid sex hormones action on male sexual behavior tend to investigate androgens and estrogens separately (199,235-238). Furthermore, steroid sex hormones may influence both gender-identity and sexual orientation (239,240), even though in humans this action is mitigated by the strong influence of psychosocial factors.

 

Estrogens and Gender Identity and Sexual Orientation

 

Testosterone aromatization to estradiol in the brain was traditionally considered the key step in the development of a male brain and in determining sexual dimorphism of the central nervous system in non-primate mammals (241-243). According to Dörner’s hypothesis (244), prenatal and perinatal brain exposure to estrogens may be responsible for the establishment of a male brain (240,245), an event occurring only in the male, but not female, brain. Accordingly, ovaries release a very small amount of estrogen, soon inactivated in rodents (4,245), while the testes produce a greater amount of androgen that is converted into estrogen. Thus, circulating estrogens are paradoxically greater in males than in females during fetal life (240,246) and this accounts for the sexual dimorphism of hypothalamic structures in rodents and other species like sheep (245-247).

 

The same mechanism seems to be also involved in the differentiation in hypothalamic structures between men and women (244,246,248). Prenatal hormonal exposure is classically considered to be involved in determining sexual orientation, on the basis of some differences in hypothalamic structures between heterosexual and homosexual men (243,248). This hypothesis is supported by the concept that brain sexual differentiation during fetal life occurs in parallel with the peak of testosterone secretion from the testis and the consequent increase in serum estradiol (240,241,243,245). Accordingly, the intrinsic pattern of mammalian brain development is female, and estrogen is required for the development of a male brain (240,243,244), thus emphasizing the role of locally produced estrogen (245). Permanent changes in the organization of different neural circuits, fundamental for sex-specific regulation of reproductive and sexual behavior, probably also occurs under the effects of estrogen (240,242,243,245,249). Considering all the above mentioned aspects, the lack of estrogen action on the developing brain in males should be considered strictly related to the direction of future development of sexual orientation, and of dimorphism of hypothalamic structures (240,241,243,245,248). Most of the data supporting this evidence, however, came from studies performed in rodents or other species, but not in humans (240,242,243,245).

 

The role of hypothalamic aromatase activity and expression in partner preference has been elegantly confirmed in rams (250). In this study, the choice of sexual partner was associated with both the volume of the ovine sexually dimorphic nucleus and different patterns of aromatase expression (250). This provides the first demonstration that differences in aromatase expression within the brain are related to partner choice and to the determination of adult sexual behavior (245,247,250). However, in humans, a clear cause-effect relationship between prenatal exposure to sex steroids (especially estrogens) and the differences in volume of some dimorphic brain areas (e.g. sexually dimorphic nucleus of the preoptic area and the intermediate nucleus) has not been demonstrated (240).

 

Aromatase-deficient men represent an interesting model to investigate the role of estradiol on human male sexual development and behavior from fetal life through adulthood (4,55,135). All men with aromatase deficiency who underwent a comprehensive evaluation of sexual behavior had male gender-identity and heterosexual orientation (4,20-22,135,138,143,144,150,152,153,156) (Table 7). The fact that congenital aromatase deficiency does not affect psychosexual orientation and gender-identity in humans suggests that estrogens do not mediate the organizational effects on male sexuality induced by early exposure to androgens. Differently from animals, psychological and social factors are the most relevant determinants of gender role behavior in men, with hormones probably having a minor role compared to other species (4,135,228,247). Accordingly, the prenatal exposure to DES, a potent estrogenic compound, is able to modify partner preference in animal studies, but not in humans (251).

 

In conclusion, aromatase plays a key role in controlling male reproductive behavior especially in animals (rodents and rams), by modulating organizational effects on the developing brain during fetal life (249,252); the latter are mediated by estrogen production within the brain and exposure to circulating estrogens. However, differences among species could explain the essential role of aromatization in rodents, rams, and monkeys (247,252,253) and its poor or minor effect in humans (4,135,153,156) and other primates, respectively (253). Thus the debate about nurture (254) versus nature (246) remains still open in humans.

 

Estrogens and Sexual Behavior

 

In adult men sexual behavior is partially dependent on testosterone, the main hormone involved in male sexuality (226,229,230). Accordingly, testosterone deficiency frequently causes loss of libido and erectile dysfunction (226,230,255). These are restored by testosterone replacement therapy, which is effective in increasing sexual interest and improving sexual function (226,227,255,256). Other hormones, however, are involved in the control of male sexual behavior, including estrogens (257,258).

 

In experimental animal models, the knockout of estrogen pathways or a pharmacologically induced estrogen deficiency results in severe impairment of sexual behavior (4,16,26). Accordingly, ArKO mice (259), αβERKO male mice (260) and αERKO mice (16,109) all exhibit a significant reduction in mounting frequency and prolonged latency to mount when compared with wild-type animals (16,26). On the contrary, βERKO mice did not show abnormalities of sexual behavior (16,112). These findings suggest that androgen receptor activation alone is not sufficient for fully normal sexual behavior in rodents and that a normal functioning ERα together with adequate levels of circulating or locally produced estrogen are required for mounting behavior in male mice (4,55).

 

Less is known about the role of estrogens in sexual behavior in men since the relative importance of testosterone and its metabolite estradiol on male sexual behavior is still not known. In the past five years, only a few studies have investigated the direct effect of estrogen on male sexual behavior (261,262), indirect evidence being available only from rare cases of men with congenital estrogen deficiency (4,55,130,135,143,144) (Table 7). A detailed sexual investigation of aromatase-deficient men documented an increase in all the parameters of sexual activity during estrogen treatment (153,156), with the best outcome in terms of sexual behavior obtained only when a concomitant normalization of both serum testosterone and estradiol was reached (156). These results support the concept that both sex steroids are required for normal sexual behavior in men. Outside the context of congenital lack of estrogens, it is difficult to reach conclusive information on the role of estrogen on male sexual behavior because of the inadequacy of studies and the conflicting results reported in the literature.

 

Table 7. Sexual Behavior in Men with Congenital Estrogen Deficiency

Subjects

Authors

Sexual function

Gender identity

Psychosexual orientation

Estrogen Resistance

(Age:28 years)

Smith et al.1994(19)

Libido: normal.

Morning erections: normal.

Nocturnal emissions: normal.

Ejaculations: normal.

Male

Heterosexual

Aromatase Deficiency

(Age 24 years)

Morishima et al. 1995(20); Bilezikian et al.1998(136)

Libido: modest. Morning erections: normal.

Nocturnal emissions: normal.

Ejaculations: normal.

Male

Heterosexual

Aromatase Deficiency*

 (Age 38 years)

Carani et al.1997(21); Carani et al.1999(153)

Libido: normal.

Morning erections: normal.

Ejaculations: normal.

Male

Heterosexual

Aromatase Deficiency*

 (Age 28 years)

Maffei et al.2004(150); Carani et al.2005(156)

Morning erections: normal.

Libido and sexual activity have not been investigated according to the religious thinking of the patient.

Male

Heterosexual

Aromatase Deficiency

 (Age 27 years)

Herrmann et al. 2002(138); Herrmann et al. 2005(142)

Libido: normal.

Morning erections: normal.

Ejaculations: normal.

Male

Heterosexual

Aromatase Deficiency

 (Age 25 years)

Maffei et al.2007(150); Zirilli et al.2009(160)

Libido: normal.

Morning erections: normal.

Ejaculations: normal.

Male

Heterosexual

Aromatase Deficiency

 (Age 27 years)

Lanfranco et al. 2008(152)

Libido: normal.

Morning erections: normal.

Ejaculations: mild praecox ejaculation.

Male

Heterosexual

Aromatase Deficiency

 (Age 27 years)

Baykan et al.2013(143)

No sexual dysfunction reported before and during treatment

Not reported

Not reported

Aromatase Deficiency

3 men

 (26-44 years)

Pignatti et al.2013(144)

No sexual dysfunction reported before and during treatment

Not reported

Not reported

Aromatase Deficiency

 (Age 24 years)

Chen et al.2015(146)

Libido: normal.

No sexual dysfunction reported

 

Not reported

Not reported

Aromatase Deficiency

 (Age 25 years)

Miedlich et al.2016(147)

Libido: normal.

Morning erections: normal.

 

Not reported

Not reported

* Only these two patients underwent an extensive, well-designed study of sexual behavior in terms of psychosexual issues (gender identity and sexual orientation) and sexual function (desire and erectile function), while for other patients the information was obtained by patients’ interview and medical history. No data on sexual behavior are available for the other patient with estrogen resistance (133) and for the other aromatase deficient-men (139,140,145,148).

 

Recently, a very elegant study provided evidence-based information on the relative role of testosterone and estradiol on male sexual function in men (199). In this study, a considerable number of healthy men (n= 400) underwent gonadotropin suppression by the administration of a GnRH analogue (goserelin acetate), resulting in testosterone and estradiol suppression (199). In order to investigate the placebo effect and the effects of testosterone and estradiol, subjects were assigned to receive i) placebo, ii) testosterone treatment at different dosages, and iii) testosterone at different dosages plus the aromatase inhibitor anastrozole (199).

 

In the testosterone group, serum testosterone and estradiol varied from physiological levels to low levels according to the different doses of exogenous testosterone in each group and the estradiol to testosterone ratio remaining substantially unchanged in all groups (199). This pharmacologic scheme allowed testing the effects of lowering both serum testosterone and estradiol in a similar way on several physiological functions; the result was a decline of both sexual desire and erectile function in parallel with the decrease of both sex steroids (199). In the testosterone plus anastrozole group, the decline of serum testosterone paralleled that obtained in the testosterone group, while serum estradiol was quite suppressed and changed to a lesser degree in each testosterone dose group, thus fluctuating across very low values (199). In this group, both sexual desire and erectile function were severely affected in patients with low serum levels of estradiol despite normal serum testosterone in patients taking the higher doses of testosterone (199). Conversely, goserelin treatment resulted in the maximum reduction of both sexual desire and erectile function in the placebo group (199). These results confirm observations in aromatase-deficient men (135,153,156) and suggest that estrogen deficiency is largely responsible for the impairment in sexual function occurring when serum testosterone is suppressed in hypogonadal men (199). A possible role of estrogen on male sexual function is also provided by further studies showing that testosterone therapy is more effective on libido when the treatment produces serum estradiol levels greater than 5 ng/dL (235) and that this serum estradiol is directly related to sexual function in men (236). In particular, serum estradiol is associated with sexual activity and desire, but not with erectile function (263). In addition, exogenous estradiol improves sexual desire in men with low testosterone and prostate cancer (264).

 

Notwithstanding these studies, the role of estrogen in male sexual behavior remains controversial (257) since several studies reached opposite conclusions. In particular, Sartorius et al. found that DHT was effective in maintaining male sexual function in healthy, older men, despite its suppressive effect on both testosterone and estradiol, suggesting that male sexual function can be ensured without aromatization (237). Furthermore, other cross-sectional studies failed to demonstrate a clear association between serum estradiol and male sexual function (238,265).

 

To add further complexity, estrogen action on erectile function seems to be biphasic, since estrogen deficiency may affect the ability to achieve an erection, yet estrogen excess and an increased estradiol to testosterone ratio is associated with an impaired erectile function. A Chinese group reported that serum estradiol is higher in a large sample of adult men with erectile dysfunction compared to men with normal erectile function, while it was not different among men with and without premature ejaculation (266,267). The same group has proposed that high serum estradiol and reduced estradiol to testosterone ratio may be independent risk factor for organic erectile dysfunction (268). Similar results have been obtained by other authors in hypogonadal men where high serum estradiol was associated to more severe erectile dysfunction (269). Also in an experimental rabbit model of the metabolic syndrome (a model used to investigate erectile function), erectile dysfunction is associated with high serum estradiol rather than low testosterone in line with the above mentioned clinical data (270). Conversely, other authors did not find any correlation between testosterone to estradiol ratio and erectile dysfunction (271). All these data, however, needs to be confirmed by further studies since the strength of evidence is weak due to many flaws such as the lack of data on the cause of sexual dysfunction, the poor accuracy of estradiol assay, and the retrospective collection of data.

 

A possible explanation for these results is that a serum estradiol in the normal male range is required for a fully normal male sexual function in addition to testosterone, while both estrogen deficiency and estrogen excess have a negative impact on male sexual activity (156,236,238).

 

Finally, estrogen receptors and the aromatase enzyme have been identified in the penile tissue of a large number of species, including humans (272-274) suggesting direct estrogenic activity within the penis. At present, knowledge on estrogen action within the penis derives from the observation that: i) male offspring exposure to estrogen-like endocrine disruptors in utero induces micropenis and hypospadias (176), and that ii) penile development and function is estrogen-dependent in animals (275).

 

OTHER NON-REPRODUCTIVE PHYSIOLOGICAL ESTROGEN ACTIONS IN MEN

 

Estrogens, Metabolism, and Cardiovascular Diseases in Men

 

The role of estrogen on glucose and insulin metabolism in men is difficult to establish since it is challenging to differentiate androgen from estrogen actions in vivo. In estrogen-deficient men, both insulin resistance and fasting glucose are increased and improve during estrogen treatment (150,158,161), confirming data from mice models (16). Thus, severe estrogen to testosterone ratio imbalance (increased androgens and decreased estrogens) seems to favor  the development of insulin resistance in men (150,151,158), and not only in estrogen-deficient men (276). In healthy men, the administration of an aromatase inhibitor in a double blind, randomized, controlled, crossover study led to a decrease of insulin sensitivity (277). Several other clinical studies confirmed that aromatase inhibition worsens insulin sensitivity in older men (278), and in obese men, independently from the increase in serum testosterone (279). The same results come from studies comparing exogenous estradiol to GnRH analogues for the treatment of prostate cancer showing that fasting both fasting glucose and cholesterol decreased in men treated with estradiol (224).

 

In accordance with this findings, relative estrogen deficiency is found in men with type 2 diabetes mellitus and low serum testosterone who display also low serum estradiol (280).

 

Furthermore, congenital estrogen deficiency is associated with an altered lipid profile (22,161), mainly characterized by higher total cholesterol and triglycerides serum levels, higher low-density lipoprotein (LDL) cholesterol and very low high-density lipoprotein (HDL) cholesterol (11,135). In these patients, estradiol treatment induces a moderate increase of HDL-cholesterol together with a small reduction of triglycerides, total cholesterol, and LDL cholesterol (21,135,138,150), resembling the effects of estrogen on lipid metabolism exerted in females (10). The administration of aromatase inhibitors leads to no change in serum lipids after 12 months of therapy (278). Outside the context of rare congenital diseases, the effects of estrogens and antiestrogens on serum lipids remains not well established due to conflicting results, and additional studies are needed (24).

 

In community-based men aged 20-70 years, high serum E2 is associated with reduced carotid plaque and serum E2 is directly related to carotid intima media thickness (281). The T to E2 ratio was associated with increased atheromatous plaque inflammation and increased risk of subsequent major adverse cardiovascular events (MACE) in men with documented atherosclerotic disease (282). In an aromatase-deficient man the normalization of serum E2 induced by estradiol replacement was able to reduce carotid atherosclerotic plaque volume on ultrasonography (150). While serum E2 may exert protective effects against atherosclerosis, the serum T to E2 ratio maybe a marker of low serum T and increased adiposity, especially in overweight and obese men and may be associated with increased cardiovascular (CV) risk (282,283). However, these clinical studies suffer from serum E2 measurement with commercially available immunoassay that are unreliable for E2 values in the normal male serum range (41,42,131). An association between coronary artery calcification (a surrogate radiological markers of coronary atherosclerosis) and both low serum testosterone and low serum estradiol measured by liquid chromatography/tandem mass spectrometry (LC-MS/MS) has been found in men participating to the Framingham Heart Study (284).

 

In hypogonadal men, estrogen deficiency, but not testosterone deficiency is responsible for vasomotor symptoms (i.e., hot flushes). In men treated with an LHRH analogue and replaced with placebo, different dosages of T alone or T plus aromatase inhibitors, only estrogen deficiency resulted in the occurrence of vasomotor symptoms (285). The main role of estrogen deficiency in the occurrence of hot flushes has been also confirmed in men with prostate cancer treated with exogenous estradiol or LHRH analogue, being hot flushes significantly more prevalent in men with estrogen deficiency due to LHRH therapy. Taken together these results reinforce the concept that in men with hypogonadism several clinical manifestations are due to relative estrogen deficiency rather than to testosterone deficiency per se (285).

 

Estrogens and the Male Bone

 

There is increasing evidence suggesting that circulating estrogens plays a key role in bone health in men, as in women (286). The relative contribution of androgen versus estrogens in the regulation of the male skeleton, however, is complex and relatively unclear (287). Some estrogen actions on male bone, such as bone maturation and the acceleration of growth arrest, are now well defined (286,288). The important role of estrogen in bone metabolism in men has been characterized in the last 15 years by means of the description of rare case reports of estrogen-deficient men (135,152) and by several epidemiological studies (289,290). All patients with congenital estrogen deficiency due to estrogen resistance (19,133,134) or aromatase deficiency (20,136-149). have unfused epiphyses in adulthood and fail to reach their closure and complete bone maturation (11,18,25,130,286). Estrogen replacement therapy led to skeletal maturation and improvement of bone mineral density in all aromatase-deficient men described so far (11,135,144) in a dose-dependent way (147,154) while testosterone treatment did not (21,22,159). During puberty and late adolescence, epiphyseal closure, growth arrest, the achievement of peak bone mass, and final bone maturation are mainly under the control of estrogens and all these processes do not progress in the case of severe estrogen deficiency leading to tall stature and osteoporosis (21,22,135,286). The eunuchoid body proportions of the skeleton typical of hypogonadal men are the effect of estrogen deficiency during late adolescence and of the disproportional growth between long bones and the appendicular skeleton (11,286). During adulthood, both normal circulating estradiol and testosterone are required for maintaining bone mineral density in aromatase-deficient men (136,159,160) as well as in the general male population (132,286,287,289-292). Estrogen action on bone seems to be possible only when circulating estradiol is above a threshold between approximately 15 and 25 pg/mL (55-92 pmol/L) (286,289). This mechanism has been suggested both for growth arrest and bone maturation (152) and for bone mineral density (BMD) (132,289,292) and suggest that circulating estrogens above this threshold are required for optimal skeletal maturation and mineralization in men (130,286). Relative estrogen deficiency, rather than testosterone deficiency, is responsible for bone loss in hypogonadal men as clearly demonstrated when using different doses of exogenous testosterone alone or in combination with a potent aromatase inhibitor in men treated with GnRH analogues (293). In this study, BMD decreased and indices of bone resorption increased only in the group of men treated with both testosterone and anastrozole independently from the dose of exogenous testosterone administered to men with pharmacologically-induced hypogonadism (293). Furthermore, this study confirms that serum estradiol below 10 pg/mL is particularly harmful for bone health (293).

 

Similarly, in men with prostate cancer, estrogen deficiency induced by Androgen Deprivation Therapy (ADT) is the main factor involved in bone loss, increase of bone fragility, and the occurrence of osteoporotic fractures since serum estradiol falls below 5 pg/mL in men receiving ADT (294-296) similar to what happens in women taking antiestrogens for breast cancer (132,292,297). Accordingly, new strategies for the treatment of prostate cancer by using blockers of the androgen receptor and exogenous estradiol are being investigated in order to mitigate the risk of bone fractures and other side effects of ADT (224,295,296). In particular, transdermal estradiol resulted effective in preventing bone loss compared to LHRH analogue in men with prostate cancer (298).

 

Figure 6. Genetically determined factors influencing the amount of serum estrogens in men starting from a determined amount of circulating serum T.
[SNPs: single-nucleotide polymorphisms; T: testosterone; DHT: dihydrotestosterone; E2: estradiol; GnRH: gonadotropin releasing hormone; LH: luteinizing hormone; FSH: follicle-stimulating hormone]

Except for rare cases of congenital estrogen deficiency (162), clinical experimental models of estrogen deficiency (199,285,293), and ADT (295,296), the most common condition in men is relative estrogen deficiency induced by hypogonadism and low serum levels of T, the precursor for estrogen production (Figure 1) (131,290,299,300). Relative estrogen deficiency may occur in hypogonadal men (299,300) especially in those with a less functioning aromatase enzyme (290,301), whose function is mainly under genetic determination (Figure 6) (131,132,292). The association between some polymorphisms of both ERα (302,303) and aromatase (304,305) genes and low bone mineral density (BMD) have clearly pointed out that the estrogen pathway is crucial for bone health in men confirming epidemiological and clinical data. These data have been recently confirmed by studies based on genome-wide association studies. These studies provided evidence on the role of aromatase enzyme in relative estrogen deficiency occurring in men with hypogonadism and its impact on BMD (Figure 6). Accordingly, a genome-wide association study demonstrated how some genetic variants of the aromatase enzyme are correlated with circulating serum E2 and BMD and that genetically determined values of circulating estradiol are linked to BMD by estimating that every 1 pg/mL of serum E2 corresponds to a genetically determined increase of BMD of 0.048 standard deviation (306). These results have been recently replicated by another GWAS study (307). Similarly, serum estradiol but not testosterone was identified as a causal factor in bone osteoporotic fractures in 175,583 men studied using a Mendelian randomization approach (308). Thus, the lower the serum estradiol the greater the extent of bone loss in hypogonadal men, the decrease of serum T exerting a direct, minor role (131,132,157,292,293). As a matter of fact, in male to female transexuals, high serum E2 induced by estrogen therapy maintains and increases bone mass despite low serum T (309,310). The degree or relative estrogen deficiency in hypogonadal men depends on several factors, including aromatase functioning (Figure6).

 

Estrogens, Body Composition, and Obesity in Men

 

The role of sex steroids action on body composition and adipose tissue metabolism in humans is long been recognized (311). Testosterone has been largely considered the main sex steroid involved in fat pathophysiology in men (311,312). However, it has become clear that estrogen also plays an important role in adipose tissue physiology in men since the first description of human cases of congenital estrogen deficiency (11,135,151) and the generation of knock-out mice models of estrogen deficiency (16,117,313,314) showing a phenotype of increased adiposity in spite of normal to increased serum testosterone.

 

Accordingly, both aromatase enzyme and ERs are expressed in the adipose tissue and in the skeletal muscle (12,18,24,25).

 

BODY COMPOSITION

 

Testosterone increases muscle mass and reduces fat mass in vivo (312). However, it is now clear that much these effects are due to its conversion into estradiol.

 

In the context of an experimental in vivo setting, healthy younger men treated with GnRH analogues and then substituted with different doses of exogenous testosterone alone together with placebo or in combination with an aromatase inhibitor, showed an increase in both subcutaneous and visceral fat mass mainly in presence of estrogen deficiency induced by the aromatase inhibitor (199). Similar results demonstrate that fat mass increases only in the group of healthy men with relative estrogen deficiency (below 15 pg/mL) induced pharmacologically by the short-term administration of GnRH analogue and aromatase inhibitors, but not in men with estradiol in the normal range (315). In accordance obese men treated with supraphysiological doses of transdermal testosterone or dutasteride showed an increase in fat mass only in the group taking an aromatase inhibitor compared to placebo (279).

 

Similarly, men treated with androgen deprivation therapy for prostate cancer increase their fat mass and weight and loose muscle mass (316-318). However, a double-blind randomized study tested the effects of 6 months of transdermal estradiol therapy in men on androgen deprivation therapy, and this study showed no beneficial effect of estradiol to prevent the increase of fat mass (319). This study was prematurely stopped due to Covid 19 pandemic, and the premature discontinuation might have resulted in insufficient power to detect a benefit (319).  Estradiol therapy also seems to not influence fat-free mass and muscle size (199).

 

Congenital human models of sex steroids deficiency also support the importance of estrogen deficiency in the body fat mass increase since men with congenital estrogen deficiency show increased adiposity (11,135,150,151) and patients with complete androgen insensitivity syndrome do not accumulate fat mass even in  absence of testosterone action thanks to a normal production of estrogens (320).  The mechanisms through which estrogens modulate fat mass in men are not fully understood and several, different actions are involved and interlinked in a complex network.

 

ESTROGENS AND THE GH/IGF-1 AXIS

 

Estrogens modulate the GH/IGF-1 axis by enhancing both the GH and IGF-1 secretion in men (321). Thus, estrogen deficiency indirectly leads to body composition changes related to the inhibition of the GH-IGF-1 axis (321).

 

ESTROGEN ACTION ON ADIPOSE TISSUE

 

It is well known that the expression of the ER within adipose tissue is decreased in obese women (322) and that the deletion of the ERS1 gene encoding for the ER in adipocytes increases the adipocytes volume and total fat mass in both male and female rodents (323), but the underlying mechanisms needs to be fully ascertained.

 

Recently, the study performed in approximately 700 women and 800 men demonstrates that the expression of the estrogen receptor ESR1 within adipose tissue is inversely associated with abdominal fat mass and insulin insensitivity (324). Therefore, men with relative estrogen deficiency due to lower ESR1 expression in adipose tissue or low circulating estradiol, or both, tend to have higher fat mass and insulin resistance, thus pointing out on a main role of estrogens on weight and body composition in men.  The reduction of ESR1 expression was associated with mitochondrial dysfunction of adipocytes, and these effects seem to be mediated by the reduction of expression of Polg1, a subunit of the polymerase enzyme involved in mitochondrial DNA replication and transcription (324). Reduced ESR1 expression in both white and brown adipocytes of ERKO mice (with deleted ESR1 gene) leads to increased fat in the former and reduced energy burning in the latter (324). Accordingly, in healthy adults of both sexes, serum E2 regulates brown adipose tissue thermogenesis, the latter being increased in women compared to men and associated with serum E2 (325,326).

 

ESTROGENS AND FEEDING BEHAVIOR

 

Sex hormones are able to influence adiposity in both men and women not only through peripheral mechanisms but also through a direct action on the brain (326,327) where they may modulate both energy balance and feeding behavior (328,329). Several data suggest that estrogens modulate energy balance at the hypothalamic level where they exert an anorexigenic action (326).

 

In addition to the energy expenditure, increasing evidence suggests that estrogens modulate appetitive behavior and food intake through their action on hedonic pathways operating at central level. Estrogens seem to lessen appetite and to reduce food intake. Recent studies have focused on the role of locally produced estrogens within the amygdala where its amount seems to be crucial for the regulation of feeding behavior. The amygdala is involved in the control of food intake both in humans and animals. In particular, functional imaging studies have demonstrated the activation of amygdala when images/videos of food or eating behavior are administered to volunteers and that functional dysregulation of this kind of activation is present in obese men and women, especially in hunger conditions (330). By measuring aromatase availability in the amygdala using positron emission tomography (PET) with the aromatase inhibitor [11C]vorozole in normal- weight, overweight, or obese men, Biegon et al. demonstrated that aromatase availability in the amygdala, but not circulating sex steroids, was inversely correlated to BMI (331). In particular, the aromatase inhibitor [11C]vorozole (a surrogate marker of aromatase expression/activity in vivo and of locally produced estrogens) was less available within the amygdala of obese men compared to healthy and normal-weight men (331). This suggests that locally produced estrogens within the amygdala can suppress eating behavior thus contributing to the modulation of weight gain since they facilitate the individual control of impulsive eating through cognitive/hedonic central effects within the brain (331).

 

Several actions of estrogens on food intake, energy homeostasis and body fat mass have been well characterized mainly in women (326) and till now the current knowledge was that estrogens are less important in men than in women in the control of these physiological functions, but recent evidence is changing this paradigm. In fact, all of the above findings point to an important role of estrogens on body composition, energy expenditure and control of feeding behavior in men and suggest that estrogen may represent a possible target to prevent and/or reverse weight gain  in men (332)

 

Other Non-Reproductive Estrogen Functions in Men

 

During the last five years an increasing body of evidence suggest that estrogen may play a role on several other non-reproductive function in men. Among them the most investigated are cognitive function (333) and aging (334,335). However, at present more evidence is needed to confirm the putative role of estrogens on these functions in men.

 

EFFECTS OF ESTROGEN EXCESS

 

Effects of Exposure to Excess Estrogens in Animals

 

In order to evaluate the effect of estrogen excess on the reproductive tract, several studies have been performed in various animal species treated with diethylstilbestrol (DES), a synthetic, potent estrogenic compound (336). The period between 13 to 24 weeks of human fetal life corresponds with the highest susceptibility of male reproductive organs to endocrine disruptors (4,53,55,336). Many studies in rodents suggest that the inappropriate exposure to estrogen in utero and/or during the neonatal period impairs the hypothalamic-pituitary-gonadal axis, testicular descent, efferent ductule function and testicular function (26,31,176,178). The latter effect is a direct consequence of the exposure to estrogen excess, of the indirect effect of perturbations in circulating hormones, and of the ability of the efferent ductules to reabsorb fluid. It seems that ERβ may mediate the process through which excess estrogens produce negative effects on male reproduction (26,31,57). The effects of estrogen excess during the neonatal period can induce irreversible alterations of the testis that become manifest in adulthood, consisting of permanent changes in both testis function and spermatogenesis (26,31).

 

AROMATASE OVER EXPRESSION IN RODENTS

 

The transgenic model of mice overexpressing the aromatase enzyme (AROM+) exhibits highly elevated serum estradiol concentrations together with a decrease of serum testosterone levels due to gonadotropin suppression (211,337). The phenotypic abnormalities of AROM+ males are like those developed by mice that are perinatally exposed to estrogens. The most frequent abnormalities include: undescended testes, testicular interstitial cell hyperplasia, hypoandrogenism, and growth inhibition of accessory sex glands (211). The impairment of spermatogenesis observed in AROM+ may be due to multiple factors, including cryptorchidism, abnormal Leydig cell function, testosterone deficiency or hyperestrogenemia (211). Thus, estrogens are thought to inhibit Leydig cell development, growth and function, resulting in the final suppression of androgen production (26). Furthermore, the observation of numerous degenerating germ cells and the absence of spermatids within the seminiferous tubules of AROM+ mice suggest that germ cell development arrests at the pachytene spermatocyte stage (26). However, a possible role of cryptorchidism per se on germ cell arrest cannot be excluded since cryptorchidism is known to induce germ cell arrest in rodents (338). Interestingly, the spermatogenic arrest occurred at a stage where P450arom expression is generally high. The spermatogenic arrest found in the AROM+ mice could be explained, at least in part, by the suppression of FSH action (211,337). In fact, the reduced serum FSH levels associated with normal LH levels provide further evidence of the inhibiting actions of estrogens on FSH secretion in in AROM+ males (211,337).

 

Effects of Exposure to Excess Estrogens in Men

 

The observation that the clinical use of DES by pregnant women to prevent miscarriage is associated with a dramatic increase in the incidence of genital malformations in their sons represents the first evidence in humans on the potential for estrogen excess to provoke urogenital malformations (339). The most frequent structural and functional abnormalities include epididymal cysts, meatal stenosis, hypospadias, cryptorchidism and microphallus (339-341). The frequency of abnormalities is dependent on the timing of estrogen exposure; in fact, men who were exposed to DES before the 11th week of gestation (i.e. the time of Műllerian ducts formation) had a two-fold higher rate of abnormalities than those who were exposed later (339,341). These data support the hypothesis that the asynchrony between formation and regression of embryonic reproductive structures is probably strongly influenced by estrogen exposure.

 

Various reports demonstrated that semen quality of men exposed to DES in utero is significantly worse than in unexposed controls (342), even though sperm concentrations of most of these patients was average, with normal fertility (14). The implications for human spermatogenesis in terms of exposure to environmental estrogens remain less clear. The risk of testicular cancer among men exposed to DES in utero has been a controversial issue and several meta-analyses showed a doubling of testicular cancer risk, together with increased incidences of cryptorchidism, hypospadias, and impaired spermatogenesis (343). However, more direct evidence will be necessary in order to fully understand this issue and particularly to identify the exact estrogenic mechanism of action (343). It is clear that exogenous estrogens could interfere with the development of genital structures if administered during early organogenesis (341). The main effect is an impairment of gonadotropin secretion and the imbalance of estrogen to androgen ratio, which may account for impaired androgen receptor stimulation or inhibition according to the dose, the cell type and the timing of exposure (339,341). Furthermore, it seems that an excess of environmental estrogens could be a possible cause of impaired fertility in humans (176,177,341) since environmental estrogens are associated with an increased risk of subfertility in several studies (344). Although controversial, a proposed progressive decline in sperm count has been reported in some Western countries during the past 50 years, and has been suggested to involve  negative effects of environmental contaminants, especially xenoestrogens, on male reproductive function(13,176,339,344).

 

In adult men the effects of estrogen excess are limited to rare causes of congenital aromatase overexpression and other rare conditions such as male to female transexuals taking exogenous estrogens.

 

AROMATASE OVER EXPRESSION IN HUMANS

 

Aromatase over-expression causes an increased conversion of androgens to estrogens with a consequent excess of estrogen. Excess estrogen in boys causes gynecomastia, a premature growth spurt, early fusion of epiphyses, and decreased adult height (162,345). Increased extraglandular aromatization was firstly reported in an adopted boy with prepubertal gynecomastia in 1977 (346). Four families were then described, in which several members had estrogen excess (manifested as gynecomastia in boys and men and premature thelarche in girls) due to increased extraglandular aromatization (347-349), and one case with a gain-of-function mutation of the aromatase gene (345). The latter seemed to be an autosomal dominant inherited disease (345,348). In adult men, elevated serum estradiol levels induce mild hypogonadotropic hypogonadism due to enhanced negative feedback on pituitary gonadotropins exerted by estrogens (162,345,348). This inhibitory effect of estrogen on reproductive function appears to be milder in males with aromatase excess syndrome than in patients receiving exogenous estrogens or having estrogen-secreting tumors, probably because serum estradiol and/or estrone levels are lower in the former (348). External genitalia in adult men with aromatase excess syndrome are characterized by normal penile and testicular size (162,345,348). This clinical reproductive phenotype has been observed also in other patients with aromatase excess syndrome due to gain-of-function mutations of the aromatase enzyme (162,350-352). Even though spermatogenesis and sexual behavior were not specifically studied, the adult men described were fertile and reported normal libido (345,348) and sperm count was normal in other studies (350). In these patients, treatment with an aromatase inhibitor reduces estrogen levels and normalized testosterone, LH and FSH serum levels (345,353), confirming a crucial role of estrogen in the suppression of both gonadotropins in men.

 

ESTROGEN EXCESS IN ADULT MEN

 

Klinefelter’s Syndrome has been classically considered a feminizing syndrome on the basis of signs (gynecomastia) and the observation of circulating estradiol higher than normal (354,355). In the literature, however, the data concerning hyperestrogenism in Klinefelter patients are not solid since they come from single case reports or studies using old assays for the measurement of serum estradiol. Data from mouse models of Klinefelter’s are not conclusive about the real increase of circulating estrogens and aromatase expression and activity (356). Infertility in these patients is mainly due to the genetic abnormalities rather than to the hormonal status (357). However, preliminary results from a recent meta-analysis does not confirm that is higher serum estradiol in Klinefelter’s patients compared with non-Klinefelter’s men, but show a condition of relative hyperestrogenism consisting with a slightly elevated estradiol to testosterone ratio in Klinefelter’s (358).

 

Most male to female transexuals who undergo exogenous estrogen therapy continue to have sperm production and spermatogenesis progresses even after a long period of therapy with estrogens. Histological analysis of the testes removed as part of gender affirmation procedures in 72 male to female transexuals on long -term estrogen therapy (>1 year) shows the presence of germ cells and spermatids in about 80% and 40% of cases, respectively; these percentages being inversely related to testes volume (359). In particular, a reduced diameter of seminiferous tubules, Sertoli and Leydig cells abnormalities consisting with glycoproteins accumulation, germ cells and Leydig cells hypoplasia, and down regulation of Era expression in the seminiferous tubules have all been found in testes of male to female transexuals under long-term estrogen therapy (360,361). All these changes may be associated to impaired spermatogenesis ranging from the absence of spermatozoa production (362) to various degrees of reduction in number of spermatozoa and spermatozoa precursors (spermatids) in the seminiferous tubules (360,361,363). Serum T suppression below 50 ng/dL results almost constantly in a complete suppression of spermatogenesis (362), thus incomplete suppression of spermatogenesis may be considered a marker of inadequate hormonal treatment due to underdosage or lack of patients’ adherence to therapy (360,363).

 

CLINICAL IMPLICATIONS OF ESTROGENS IN MALES

 

Diagnostic Aspects: Significance of Serum Estradiol in Men

 

Approximately 50 μg of estradiol are produced daily: about 5-10μg in the testis (10 to 20%) and the remaining 40-45 μg (80 to 90%) in peripheral tissues (adipose tissue, muscle, breast, brain liver and bone) in which the aromatase enzyme is expressed (4,130,131). In adult men, the normal range of serum estradiol is around 14-43 pg/mL (51-157 pmol/L), accordingly to different studies (300,364). Based on chromatography techniques, such as liquid chromatography-tandem mass spectrometry, progress has been made in the measurement of serum estrogens within the low and low-normal range of men thereby overcoming the unreliability of immunometric commercially available assays (44). In clinical practice, the measurement of serum E2 in men is mandatory when a congenital condition of estrogen deficiency is suspected (135,162). In particular, the clinical work-up for the evaluation of male infertility may involve the serum estradiol assay when clinical aspects suggestive for aromatase deficiency, coupled with normal to high testosterone and gonadotropins levels and/or history of cryptorchidism are present (Table 6) (365). Outside this clinical context, the measurement of serum E2 could be helpful to identify a condition of relative estrogen deficiency in men with hypogonadism and osteoporosis and hot flushes. However,  the accuracy of most of the major commercially available kits for the detection of serum estradiol remains poor, especially for low serum levels of estradiol typical of the male range (1,41,42,366,367) leaving the measurement of serum E2 in men substantially not indicated in the clinic (132,292,297,358). Therefore, the assay of serum estradiol is suggested only if the method used in clinical laboratories has a very high sensitivity and specificity (e.g. 3rd generation RIA or some immunometric assays with an acceptable accuracy) (290,301,368-370). At present, the gold standard test for E2 measurement remains the gas chromatography/tandem mass spectrometry (41-44,366,367) and its progressive introduction in laboratories for clinical routine evaluations of sex steroids in recent years (1,42-44,371,372) allows precise and accurate sex steroids measurement in a clinical setting (371,372) allowing ruling in/ruling out relative estrogen deficiency in men (131,132,292) and keeping serum E2 in the normal range in hypogonadal men treated with testosterone (373,374). Recently, the results from the Testosterone Trials have pointed out the importance of serum estradiol for the outcomes of testosterone replacement therapy (375). Accordingly, changes in serum estradiol  best predicted not only BMD increase (an expected result) but also other classical outcomes of testosterone therapy in hypogonadal men such as sexual desire, hemoglobin, and HDL cholesterol suggesting that serum estradiol assayed by LC-MS/MS may be a good clinical marker of adequate testosterone substitution (375).

 

Estrogens and Male Infertility: Clinical and Therapeutic Implications

 

Estrogens are involved in male fertility and could represent a potential factor involved in the pathogenesis of infertility as well as a possible pathway to explore new therapies for human male infertility.

 

ESTROGEN TREATMENT

 

At present there is no indication to prescribe estrogen compounds to men, except for the treatment of rare diseases such as congenital estrogen deficiency (130,135,365) or in the management of transgender patients. The increasing evidence of the existence of several testosterone actions that are mainly mediated by estrogens theoretically support the concept that tailoring estrogens in the treatment of hypogonadal men may improve the outcome in terms of benefits for patients (197,198). However, at present, there is no evidence on the effectiveness and safety of such therapeutic strategy. In the future, advances in the field of routine clinical measurement of very low amounts of circulating estrogens (1,42-44,371) will open new frontiers for testing the effect of estrogen compound or of SERMs alone or combined to androgens in men with documented mild estrogen deficiency.

 

ESTROGEN TREATMENT OF AROMATASE DEFICIENT MEN

 

The clinical features common to all aromatase-deficient men are: tall stature, delayed bone maturation, osteopenia/osteoporosis, eunuchoid skeleton, bone pain, and progressive genu valgum (11,131,135,286). Estrogen replacement treatment, at the daily dose of 0.22 to 0.35 μg/kg of transdermal estradiol in adult men, should be started as soon as the diagnosis of estrogen deficiency has been reached (131,135,365). When the diagnosis is available at birth, or is achieved during infancy, low dosages of exogenous estradiol should be administered at the beginning of puberty (0.8 to 0.12 μg/kg daily) (135,141). The main target of estrogen replacement therapy in these patients is the skeleton in order to promote epiphyseal closure, bone maturation and mineralization and the completion of these physiological processes on time. Accordingly, high doses of estrogen in adult men with aromatase deficiency might be used to lead a rapid completion of skeletal maturation within 6-9 months in adults with epiphyseal cartilages still open, through rapid bone elongation and an increase in height followed by quick epiphyseal closure and growth arrest (131,135,147,154,365). Once epiphyseal closure has been achieved, estrogen replacement treatment should be continued lifelong. The main goal is to prevent bone loss and to reduce the risk of cardiovascular disease. In this case, the dose of estradiol should be reduced to ensure serum estradiol within the normal range for adult men (131,135,147,154,365). Moreover, estrogen treatment in aromatase deficient men is effective in normalizing or improving other aspects such as gonadotropin secretion, bone mineral density, glucose metabolism, insulin sensitivity, liver function, and circulating lipids (131,143,146-148,150,151,157-159). When estrogen treatment is started at puberty, the effects of estrogen treatment on spermatogenesis are unknown, but the administration of estrogens in a more physiological way could theoretically be associated with normal spermatogenesis in adulthood. Conversely in adult patients, impaired spermatogenesis is irreversible even when estradiol treatment is administered (135). Other aspects related to estrogen deficiency cannot be modified by estrogen treatment when the treatment is started during adulthood (e.g. eunuchoid body proportions, genu valgum, failure in reaching the bone peak mass, normal body weight restoration) (135,151).

 

Finally, the real impact of estrogen treatment on sexual behavior in adult aromatase-deficient men remains to be determined (135).

 

ANTI-ESTROGEN TREATMENT IN MEN

 

As estrogens act on gonadotropic feedback inhibition (157,187,190), they could be a good target in the clinical management of male infertility. The rationale is to employ anti-estrogen drugs in order to modulate gonadotropin feedback by blocking the inhibitory effect exerted by estrogen on gonadotropins and to increase both LH and FSH. This will result in increased testosterone and FSH with potential benefits on spermatogenesis (376,377). However, the real effectiveness of this approach in treating male infertility remains to be established, since conflicting results are available (4,129,172,173,200,377-381) and this kind of treatment remains empirical and ‘off label‘ (200,376,378). Thus, the real efficacy of anti-estrogens is far from being elucidated and whether the increase of sperm density induced by anti-estrogens is actually related to a real improvement of both sperm fertility and pregnancy rates is a matter of debate (4,129,200,377) (Table 8).

 

Since the 1960s, anti-estrogen agents have been used as an empirical treatment of male infertility (378,382) based on their modulation of the hypothalamic-pituitary testicular axis. The main classes of drug that have been tested are aromatase inhibitors. They are the most potent blockers of the estrogen-mediated negative feedback on gonadotropins and excites LH and FSH secretion aiming to stimulate spermatogenesis (383). However, no clear evidence of direct effects of anti-estrogens on spermatogenesis exists (200,376,383), but LH and FSH serum levels generally increase during aromatase inhibitor administration in infertile men (384).

 

Clomiphene at a dosage of 25-50 mg daily for 3-12 months, or tamoxifen at dosage of 20-30 mg daily for 3-6 months, represent the most frequently used anti-estrogen agents for the treatment of male infertility (385) (Table 8); on the contrary the new generation of selective estrogen receptor modulators does not result in significant changes in male fertility (386) (Table 8).

 

Table 8. Dosages and Time Duration of Oral Anti-Estrogen and Aromatase Inhibitors Used in Male Infertility and Their Different Effects on Semen Analysis

Treatment

Dose (mg/daily)

Duration (months)

Effects on semen analysis

Anti-estrogens

 

 

 

Clomiphene

25-50

3-12

Semen volume: No effect or ↑

Total sperm number: No effect or ↑

Sperm concentration: No effect or ↑

Sperm motility: No effect or ↑

Sperm morphology: No effect or ↑

Tamoxifen

20-30

3-6

Semen volume: No effect

Total sperm number: No effect

Sperm concentration: No effect or ↑

Sperm motility: No effect

Sperm morphology: No effect

Tamoxifen

and

Testosterone undecanoate

20

120 orally

6

Semen volume: No effect

Total sperm number: ↑

Sperm concentration: No effect

Sperm motility: ↑

Sperm morphology: ↑

Aromatase inhibitors

 

 

 

Testolactone

2000

8

No effect

Testolactone or Anastrozole

100-200

6

Semen volume: ↑

Total sperm number: ↑

Sperm concentration: ↑

Sperm motility: ↑

Sperm morphology: ↑

Letrozole

2,5

6

Semen volume: No effect

Total sperm number: ↑

Sperm concentration: ↑

Sperm motility: ↑

Sperm morphology: No effect

The use of these drugs is still off-label.

 

Clomiphene (25-50 mg/day) has been recently studied in a cohort of 86 men with hypogonadism for six months (387). This treatment represented an effective and apparently safe alternative to testosterone supplementation in hypogonadal men wishing to preserve their fertility (387). Furthermore, Ghanem et al. have recently found that combined treatment with clomiphene (25 mg/day) and an antioxidant drug (vitamin E) increased the pregnancy rate and improved sperm count and progressive motility in men with idiopathic oligoasthenozoospermia (388). More or less these data have been confirmed by several studies, most of them being retrospective or observational (389-391), with few RTCs studies available (392,393). Notwithstanding the improvement of sperm parameters in a variable percentage of men with infertility (393), a recent systematic review points out a possible impairment of sperm parameters (a decrease in semen count, concentration, motility, morphology and total motile sperm count) in up to 20% of patients treated with clomiphene citrate, this impairment of sperm remaining irreversible in 17% among men who had a decline in semen parameters after therapy discontinuation (394). In men with secondary hypogonadism treated with testosterone, enclomiphene (the transisomer of clomiphene) was able to prevent gonadotropin suppression and the related oligospermia compared to placebo (395) and these preliminary data have been confirmed by other studies (396)(Table 8). Clomiphene may be used off-label, but enclomiphene has not been approved by regulatory agencies and its use is limited to experimental trials (396).

 

Tamoxifen (20 mg/day) has been also used in combination with oral testosterone undecanoate (120 mg/day) in men affected by idiopathic oligozoospermia. This combined treatment was effective in improving not only the sperm parameters (total sperm number, sperm morphology and motility), but also the pregnancy rate (397). In 2012, Moein et al. studied thirty-two azoospermic infertile men with proven non-obstructive azoospermia, administrating Tamoxifen for 3 months (398). Tamoxifen treatment led to the recovery of spermatozoa in the ejaculates of six patients (398). These studies showed that treatment of patients with non-obstructive azoospermia with anti-estrogenic drugs like tamoxifen can improve the results of sperm recovery in testis samples and also increase the chance of pregnancy by microinjection. Also other non-controlled trials suggest improvements in sperm quality or sperm concentration(399,400), however, no well-performed clinical trial has confirmed these results (376), except for one RCT comparing tamoxifen alone and tamoxifen plus folate with placebo confirming that tamoxifen increased sperm concentration in men with sperm abnormalities (399) (Table 8). A recent meta-analysis including a very small number of studies supports the empirical use of the estrogen antagonists clomiphene and tamoxifen at the dose of 50 mg and 20 to 30 mg daily based on the finding of the detection of a doubling rate of pregnancy outcome among men with idiopathic infertility (401). The uncertain role of these therapies on male fertility may be related to the fact that idiopathic oligozoospermia constitutes a group of heterogeneous disorders of which only a subgroup might respond to anti-estrogen therapy. However, studies have failed to identify the characteristics of this subgroup and thus physicians cannot distinguish potential responders and non-responders (376).

 

Few data are available on the effect of aromatase inhibitors in male infertility (Table 8). An old study failed to demonstrate the efficacy of testolactone in the treatment of idiopathic oligozoospermic infertility (384). However, when aromatase inhibitors (testolactone or anastrozole) were administered in a selected group of infertile men with abnormal baseline testosterone-to-estradiol ratio, an improvement of fertility rate was generally obtained (172). In particular, letrozole treatment improved semen parameters and estradiol to testosterone imbalance in patients with low testosterone and increased estradiol to testosterone ratio (381). In 2011, Saylam et al. treated 27 infertile, hypogonadotropic men with 2.5 mg daily of letrozole for six months, finding an improvement of both testosterone serum levels and semen parameters after treatment (173). Thus, it seems that letrozole may facilitate some improvement in infertile men with azoospermia by improving the number of sperm in the ejaculate (173). Accordingly, a further study on the effects of letrozole on sperm parameters showed that letrozole but not placebo was effective in increasing sperm count and improving sperm motility after 6 months of treatment in a small group of 46 patients (22 on letrozole; 24 on placebo) who were azoospermic or cryptozoospermic at baseline (402) (Table 8). These results have been also replicated by not controlled studies (403-407). However, positive effects of aromatase inhibitors on sperm concentration and quality comes mainly from case series, retrospective, and cross-sectional studies, thus leaving the strength of evidence concerning the aromatase inhibitors efficacy on male fertility of low grade (200,201,377,408).

 

In men with hypogonadism, antiestrogens have the advantage to be effective in increasing serum T without suppressing gonadotropins if compared with testosterone replacement therapy, thus preserving spermatogenesis (200,201,377).

 

Data concerning the safety of anti-estrogens for treatment of male infertility are scant, especially as far as long-term treatment is concerned (195,196,376,377,409-411). Safety data regarding the use of clomiphene and tamoxifen for male infertility is limited, but information available supports their safety (410,411), the latter  might be also derived indirectly from small groups of men with breast cancer (412). Conversely, more data are available on aromatase inhibitors (195,196,377). Six months of therapy with letrozole seems to not affect psychometric tests, glucose tolerance, serum circulating lipids, markers of bone turnover, and body composition, including BMD, in obese, hypogonadal men(413). In this study, however, moderate aromatase inhibition resulted in serum estradiol still within the normal male range and all the outcomes were obtained after a short period of treatment (413). In the literature, opposite results are available and suggest possible undesired effects of aromatase inhibitors, especially on metabolism and bone. Evidence exists that high-dose aromatase inhibition might lead to several side effects, especially when patients are treated for more than 12 months with an aromatase inhibitor (195,196). Both very short-term and short-term treatment with aromatase inhibitors had deleterious metabolic effects: one study demonstrated a prompt worsening of both insulin sensitivity and lipid profile in young and older men after 28 days of treatment with letrozole (414), while anastrozole reduced insulin sensitivity in healthy men after 6 weeks of treatment (338). In the case of longer treatments (with outcomes obtained after 1 year) vertebral deformities (415) and decreased BMD (173,197,416) were found in young and older men, respectively. In addition, treatment with aromatase inhibitors lowered HDL-cholesterol in peripubertal boys (417) and in both adult and older men (195,414) while data on total and LDL-cholesterol are conflicting (277,338,414). Data available from a very small subset of male patients operated on for male breast cancer and treated with anti-estrogens (most of them with tamoxifen) provides data on long-term effects and major adverse events (412). The authors concluded that side effects and major adverse events did not differ between men and women taking anti-estrogens and that cerebrovascular or coronary events, thromboembolic events (deep venous thrombosis)(418,419), depression, muscle cramps, and hot flashes might occur also in men during anti-estrogens treatment, hot flashes being the most frequent (411,412). These data, however, should be regarded with caution due to the small sample size, the lack of a control group, and the difficulties in proving a cause-effect relationship between major adverse events and the use of anti-estrogens in men. Besides, most of the studies on antiestrogens in men are based on short-term therapy, thus safety data for long-term therapy are not available.

 

As a result, it should be remarked that none of the drugs belonging to the category of anti-estrogens (i.e. clomiphene, tamoxifen, aromatase inhibitors) is approved for the indication of the treatment of male infertility by regulatory drug agencies (e.g. FDA, EMEA, TPD and TGA Regulations) nor is recommended by guidelines provided by Scientific Societies (e.g. NICE, ASA, EAA, SIAMS etc.) for use in idiopathic infertility (420,421). At present, all the data available on anti-estrogens in male infertility comes from the use of these drugs for research purposes outside the context of clinical practice. In addition, none of the studies are of adequate design, strength, and power. For all these reasons, their clinical use remains anecdotal and is off label (195,196,200,201).

 

In conclusion anti-estrogens, alone or in combination with testosterone, may represent a potential therapy for idiopathic oligozoospermia, however this remains an empirical off-label treatment (376,401). The data set does not yet provide sufficient evidence for these applications, but there is suggestive evidence that encourages further study (401). Further well-designed studies on adequate sample size (and homogeneous groups of men with infertility) are needed to detect their true efficacy in improving the pregnancy rate, or to identify the features of the responders.

 

FUTURE DIRECTIONS

 

Notwithstanding consistent advancements in the comprehension of estrogen role in men the pathophysiology of estrogens in males remains not fully explored and further studies are advocated. Preliminary data suggest that the decline in serum estradiol is associated with all-cause mortality in community-dwelling older men, but further investigations are needed to prove this relationship (422). In addition, the knowledge of the role of estrogen-related genetic determinants in male pathophysiology is still in its infancy. In recent years some research  based on genome-wide association studies (GWAS) have provided new insights to this field (76,306-308,423). GWAS are useful in examining unexplored areas concerning physiological and pathological actions and related genetic determinants (Figure 6). Concerning estrogens in men a recent GWAS allowed disclosing more details on the association between low serum estradiol and increased adiposity and showed that higher estradiol levels were associated with lower adiposity (423). Thus, GWAS are promising for disclosing new important aspects related to estrogen pathophysiology in men and for improving the knowledge of genetic determinants of estrogens in health and disease. Advancement in the comprehension of individual genetically determined estrogen actions (Figure 6) together with the diffusion of LC-MS/MS in clinical laboratories allowing precise measurement of estrogens in men will pave the way to better tailor the management and therapy of both estrogen deficiency and excess in the human male.

 

CONCLUSIONS

 

Sex steroids account for sexual dimorphism because they are responsible for the establishment of primary and secondary sexual characteristics, which are under the control of androgens and estrogens in male and female, respectively. Advances in the understanding of the role of estrogens in animal and human models suggest a role for this sex steroid in the reproductive function of both sexes. The fact that both estrogen excess and estrogen deficiency influence male sexual development, testis function, the hypothalamic-pituitary-testis axis, spermatogenesis and ultimately male fertility, highlight the biological importance of estrogen action in males. Thus, estrogens, not only androgens, are responsible for some crucial physiological functions in men like fertility, reproduction, and bone health. In particular, the balance of serum estradiol to testosterone ratio is likely crucial for maintaining all these functions, thus suggesting that the homeostatic equilibrium between estrogens and androgens is important for the correct functioning of several physiological systems in men (11,22,34,121,158,172,381). From an evolutionary perspective, this relevance of estrogen actions in males provides an example of the parsimony operating in biological events that are crucial for the evolution of the human species such as growth and reproduction (Figure 7).

 

This chapter has addressed the reproductive effects of estrogens in males but there are emerging roles for estrogens in non-reproductive tissues. In particular, even though testosterone has traditionally been considered the sex hormone involved in bone maturation and growth arrest in men the key role of estrogens on growth has recently been revealed.

 

Figure 7. Direct and indirect (estrogen mediated) testosterone actions.
[DHT: dihydrotestosterone, AR: androgen receptor; ERs: estrogen receptors]

A major area of uncertainty is the possible role of estrogen in boys before puberty. It is known that low levels of circulating estradiol are detected in infancy when using ultrasensitive assays, but their significance is not known (130).

 

Several lines of evidence support the view that estrogens are required for, and in part mediate, androgen actions on several tissues and organs in men (Figure 7). The progress made in the last thirty years in this field have clarified the importance of estrogen in men but leaves some issues still unsolved. In particular, estrogen actions on bone and on gonadotropin secretion are now well characterized and part of the estrogen action on spermatogenesis is known, but further evidence is needed to clarify several aspects still under debate.

 

ACKNOWLEDGMENTS

 

We are indebted to Kenneth Korach S. (National Institutes of Health, Research Triangle Park, NC, United States), Evan Simpson (Hudson Institute of Medical Research, Clayton, Australia), Laura Maffei (Buenos Aires, Argentina) for their collaboration with our group in this research field.

 

A special thanks to Marco Faustini-Fustini (Ospedale Bellaria, Bologna, Italy), Antonio Balestrieri (Ospedale Bufalini, Cesena, Italy), Antonio R.M. Granata (University of Modena and Reggio Emilia), Elisa Pignatti (University of Modena and Reggio Emilia), Fabio Lanfranco (University of Turin, Italy), and Paolo Beck-Peccoz (Univesity of Milan, Italy) for fruitful discussion and collaboration in the research area of estrogen role in the human male.

We are grateful to Bruno Madeo, MD, PhD, Chiara Diazzi, MD, PhD Lucia Zirilli, MD, PhD Daniele Santi MD, PhD (University of Modena and Reggio Emilia) for their contribution in revising some parts of the previous version of this chapter (November 24, 2016).

 

REFERENCES

 

  1. Simpson E, Santen RJ. Celebrating 75 years of oestradiol. Journal of molecular endocrinology. 2015;55(3):T1-20.
  2. Hess RA, Cooke PS. Estrogen in the male: a historical perspective. Biology of reproduction. 2018;99(1):27-44.
  3. Zondek B. Mass excretion of oestrogenic hormone in the urine of the stallion. Nature. 1934;133(3354):209-210.
  4. Rochira V, Granata AR, Madeo B, Zirilli L, Rossi G, Carani C. Estrogens in males: what have we learned in the last 10 years? Asian journal of andrology. 2005;7(1):3-20.
  5. Steinach E, Kun H. Transformation of male sex hormones into a substance with the action of a female hormone. The Lancet. 1937;230(5954):845.
  6. MacDonald PC, Madden JD, Brenner PF, Wilson JD, Siiteri PK. Origin of estrogen in normal men and in women with testicular feminization. The Journal of clinical endocrinology and metabolism. 1979;49(6):905-916.
  7. Wolff E, Ginglinger A. Sur la transformation des poulets mâles en intersexués par injection d'hormone femelle (folliculine) aux embryons: avec 16 figures dans le texte. Ed. de la Librairie" Union.
  8. Johnson KJ, Heger NE, Boekelheide K. Of mice and men (and rats): phthalate-induced fetal testis endocrine disruption is species-dependent. Toxicological sciences : an official journal of the Society of Toxicology.2012;129(2):235-248.
  9. Simpson ER, Misso M, Hewitt KN, Hill RA, Boon WC, Jones ME, Kovacic A, Zhou J, Clyne CD. Estrogen--the good, the bad, and the unexpected. Endocrine reviews. 2005;26(3):322-330.
  10. Deroo BJ, Korach KS. Estrogen receptors and human disease. The Journal of clinical investigation.2006;116(3):561-570.
  11. Zirilli L, Rochira V, Diazzi C, Caffagni G, Carani C. Human models of aromatase deficiency. The Journal of steroid biochemistry and molecular biology. 2008;109(3-5):212-218.
  12. Blakemore J, Naftolin F. Aromatase: Contributions to Physiology and Disease in Women and Men. Physiology (Bethesda, Md). 2016;31(4):258-269.
  13. Sharpe RM, Skakkebaek NE. Are oestrogens involved in falling sperm counts and disorders of the male reproductive tract? Lancet (London, England). 1993;341(8857):1392-1395.
  14. Wilcox AJ, Baird DD, Weinberg CR, Hornsby PP, Herbst AL. Fertility in men exposed prenatally to diethylstilbestrol. The New England journal of medicine. 1995;332(21):1411-1416.
  15. Hamilton KJ, Arao Y, Korach KS. Estrogen hormone physiology: reproductive findings from estrogen receptor mutant mice. Reproductive biology. 2014;14(1):3-8.
  16. Couse JF, Korach KS. Estrogen receptor null mice: what have we learned and where will they lead us? Endocrine reviews. 1999;20(3):358-417.
  17. Simpson ER, Mahendroo MS, Means GD, Kilgore MW, Hinshelwood MM, Graham-Lorence S, Amarneh B, Ito Y, Fisher CR, Michael MD, et al. Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis. Endocrine reviews. 1994;15(3):342-355.
  18. Cooke PS, Nanjappa MK, Ko C, Prins GS, Hess RA. Estrogens in Male Physiology. Physiological reviews.2017;97(3):995-1043.
  19. Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. The New England journal of medicine. 1994;331(16):1056-1061.
  20. Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K. Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. The Journal of clinical endocrinology and metabolism. 1995;80(12):3689-3698.
  21. Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, Boyd J, Korach KS, Simpson ER. Effect of testosterone and estradiol in a man with aromatase deficiency. The New England journal of medicine.1997;337(2):91-95.
  22. Faustini-Fustini M, Rochira V, Carani C. Oestrogen deficiency in men: where are we today? European journal of endocrinology / European Federation of Endocrine Societies. 1999;140(2):111-129.
  23. Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiological reviews. 2017;97(1):135-187.
  24. Russell N, Grossmann M. MECHANISMS IN ENDOCRINOLOGY: Estradiol as a male hormone. European journal of endocrinology / European Federation of Endocrine Societies. 2019;181(1):R23-r43.
  25. Hammes SR, Levin ER. Impact of estrogens in males and androgens in females. The Journal of clinical investigation. 2019;129(5):1818-1826.
  26. O'Donnell L, Robertson KM, Jones ME, Simpson ER. Estrogen and spermatogenesis. Endocrine reviews.2001;22(3):289-318.
  27. Hess RA. Estrogen in the adult male reproductive tract: a review. Reproductive biology and endocrinology : RB&E. 2003;1:52.
  28. Carreau S, Silandre D, Bourguiba S, Hamden K, Said L, Lambard S, Galeraud-Denis I, Delalande C. Estrogens and male reproduction: a new concept. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas / Sociedade Brasileira de Biofisica [et al]. 2007;40(6):761-768.
  29. Carreau S, Bouraima-Lelong H, Delalande C. Role of estrogens in spermatogenesis. Frontiers in bioscience (Elite edition). 2012;4:1-11.
  30. Schulster M, Bernie AM, Ramasamy R. The role of estradiol in male reproductive function. Asian journal of andrology. 2016;18(3):435-440.
  31. Sharpe RM. The roles of oestrogen in the male. Trends in endocrinology and metabolism: TEM. 1998;9(9):371-377.
  32. Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocrine reviews. 2011;32(1):81-151.
  33. Baker ME. What are the physiological estrogens? Steroids. 2013;78(3):337-340.
  34. Gruber CJ, Tschugguel W, Schneeberger C, Huber JC. Production and actions of estrogens. The New England journal of medicine. 2002;346(5):340-352.
  35. Stocco C. Tissue physiology and pathology of aromatase. Steroids. 2012;77(1-2):27-35.
  36. Ghayee HK, Auchus RJ. Clinical implications of androgen synthesis via 5alpha-reduced precursors. Endocrine development. 2008;13:55-66.
  37. Fang H, Tong W, Shi LM, Blair R, Perkins R, Branham W, Hass BS, Xie Q, Dial SL, Moland CL, Sheehan DM. Structure-activity relationships for a large diverse set of natural, synthetic, and environmental estrogens. Chemical research in toxicology. 2001;14(3):280-294.
  38. Amir S, Shah STA, Mamoulakis C, Docea AO, Kalantzi OI, Zachariou A, Calina D, Carvalho F, Sofikitis N, Makrigiannakis A, Tsatsakis A. Endocrine Disruptors Acting on Estrogen and Androgen Pathways Cause Reproductive Disorders through Multiple Mechanisms: A Review. Int J Environ Res Public Health. 2021;18(4).
  39. Ghosh D, Griswold J, Erman M, Pangborn W. Structural basis for androgen specificity and oestrogen synthesis in human aromatase. Nature. 2009;457(7226):219-223.
  40. Lo J, Di Nardo G, Griswold J, Egbuta C, Jiang W, Gilardi G, Ghosh D. Structural basis for the functional roles of critical residues in human cytochrome p450 aromatase. Biochemistry. 2013;52(34):5821-5829.
  41. Rosner W. Free estradiol and sex hormone-binding globulin. Steroids. 2015;99(Pt A):113-116.
  42. Demers LM, Hankinson SE, Haymond S, Key T, Rosner W, Santen RJ, Stanczyk FZ, Vesper HW, Ziegler RG. Measuring Estrogen Exposure and Metabolism: Workshop Recommendations on Clinical Issues. The Journal of clinical endocrinology and metabolism. 2015;100(6):2165-2170.
  43. Santen RJ, Demers L, Ohorodnik S, Settlage J, Langecker P, Blanchett D, Goss PE, Wang S. Superiority of gas chromatography/tandem mass spectrometry assay (GC/MS/MS) for estradiol for monitoring of aromatase inhibitor therapy. Steroids. 2007;72(8):666-671.
  44. Santen RJ, Demers LM, Ziegler RG. Workshop on measuring estrogen exposure and metabolism: Summary of the presentations. Steroids. 2015;99(Pt A):1-7.
  45. Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Strom A, Treuter E, Warner M, Gustafsson JA. Estrogen receptors: how do they signal and what are their targets. Physiological reviews.2007;87(3):905-931.
  46. Fuentes N, Silveyra P. Estrogen receptor signaling mechanisms. Adv Protein Chem Struct Biol. 2019;116:135-170.
  47. Hammes SR, Levin ER. Extranuclear steroid receptors: nature and actions. Endocrine reviews. 2007;28(7):726-741.
  48. Hammes SR, Levin ER. Minireview: Recent advances in extranuclear steroid receptor actions. Endocrinology.2011;152(12):4489-4495.
  49. Prossnitz ER, Arterburn JB, Smith HO, Oprea TI, Sklar LA, Hathaway HJ. Estrogen signaling through the transmembrane G protein-coupled receptor GPR30. Annual review of physiology. 2008;70:165-190.
  50. Luconi M, Muratori M, Forti G, Baldi E. Identification and characterization of a novel functional estrogen receptor on human sperm membrane that interferes with progesterone effects. The Journal of clinical endocrinology and metabolism. 1999;84(5):1670-1678.
  51. Baldi E, Luconi M, Muratori M, Marchiani S, Tamburrino L, Forti G. Nongenomic activation of spermatozoa by steroid hormones: facts and fictions. Molecular and cellular endocrinology. 2009;308(1-2):39-46.
  52. Hess RA. Disruption of estrogen receptor signaling and similar pathways in the efferent ductules and initial segment of the epididymis. Spermatogenesis. 2014;4(2):e979103.
  53. Li X, Li H, Jia L, Li X, Rahman N. Oestrogen action and male fertility: experimental and clinical findings. Cellular and molecular life sciences : CMLS. 2015;72(20):3915-3930.
  54. Carreau S, Bouraima-Lelong H, Delalande C. Estrogens in male germ cells. Spermatogenesis. 2011;1(2):90-94.
  55. Rochira V, Balestrieri A, Madeo B, Baraldi E, Faustini-Fustini M, Granata AR, Carani C. Congenital estrogen deficiency: in search of the estrogen role in human male reproduction. Molecular and cellular endocrinology.2001;178(1-2):107-115.
  56. Luconi M, Forti G, Baldi E. Genomic and nongenomic effects of estrogens: molecular mechanisms of action and clinical implications for male reproduction. The Journal of steroid biochemistry and molecular biology.2002;80(4-5):369-381.
  57. Carreau S, de Vienne C, Galeraud-Denis I. Aromatase and estrogens in man reproduction: a review and latest advances. Advances in medical sciences. 2008;53(2):139-144.
  58. Cooke PS, Young P, Hess RA, Cunha GR. Estrogen receptor expression in developing epididymis, efferent ductules, and other male reproductive organs. Endocrinology. 1991;128(6):2874-2879.
  59. Hess RA, Gist DH, Bunick D, Lubahn DB, Farrell A, Bahr J, Cooke PS, Greene GL. Estrogen receptor (alpha and beta) expression in the excurrent ducts of the adult male rat reproductive tract. Journal of andrology.1997;18(6):602-611.
  60. Hess RA, Fernandes SA, Gomes GR, Oliveira CA, Lazari MF, Porto CS. Estrogen and its receptors in efferent ductules and epididymis. Journal of andrology. 2011;32(6):600-613.
  61. van Pelt AM, de Rooij DG, van der Burg B, van der Saag PT, Gustafsson JA, Kuiper GG. Ontogeny of estrogen receptor-beta expression in rat testis. Endocrinology. 1999;140(1):478-483.
  62. Tao K, Sun Y, Chao Y, Xing L, Leng L, Zhou D, Zhu W, Fan L. β-estradiol promotes the growth of primary human fetal spermatogonial stem cells via the induction of stem cell factor in Sertoli cells. J Assist Reprod Genet. 2021.
  63. Cripps SM, Mattiske DM, Black JR, Risbridger GP, Govers LC, Phillips TR, Pask AJ. A loss of estrogen signaling in the aromatase deficient mouse penis results in mild hypospadias. Differentiation. 2019;109:42-52.
  64. Baskin L, Cao M, Sinclair A, Li Y, Overland M, Isaacson D, Cunha GR. Androgen and estrogen receptor expression in the developing human penis and clitoris. Differentiation. 2020;111:41-59.
  65. Govers LC, Phillips TR, Mattiske DM, Rashoo N, Black JR, Sinclair A, Baskin LS, Risbridger GP, Pask AJ. A critical role for estrogen signaling in penis development. Faseb j. 2019;33(9):10383-10392.
  66. Fisher JS, Millar MR, Majdic G, Saunders PT, Fraser HM, Sharpe RM. Immunolocalisation of oestrogen receptor-alpha within the testis and excurrent ducts of the rat and marmoset monkey from perinatal life to adulthood. The Journal of endocrinology. 1997;153(3):485-495.
  67. Macheroni C, Lucas TFG, Porto CS. The role of estrogen receptors in rat Sertoli cells at different stages of development. Heliyon. 2020;6(11):e05363.
  68. Oliveira CA, Mahecha GA, Carnes K, Prins GS, Saunders PT, Franca LR, Hess RA. Differential hormonal regulation of estrogen receptors ERalpha and ERbeta and androgen receptor expression in rat efferent ductules. Reproduction (Cambridge, England). 2004;128(1):73-86.
  69. Hess RA, Bunick D, Lee KH, Bahr J, Taylor JA, Korach KS, Lubahn DB. A role for oestrogens in the male reproductive system. Nature. 1997;390(6659):509-512.
  70. Chimento A, Sirianni R, Delalande C, Silandre D, Bois C, Ando S, Maggiolini M, Carreau S, Pezzi V. 17 beta-estradiol activates rapid signaling pathways involved in rat pachytene spermatocytes apoptosis through GPR30 and ER alpha. Molecular and cellular endocrinology. 2010;320(1-2):136-144.
  71. Shughrue PJ, Lane MV, Scrimo PJ, Merchenthaler I. Comparative distribution of estrogen receptor-alpha (ER-alpha) and beta (ER-beta) mRNA in the rat pituitary, gonad, and reproductive tract. Steroids. 1998;63(10):498-504.
  72. Mowa CN, Iwanaga T. Expression of estrogen receptor-alpha and -beta mRNAs in the male reproductive system of the rat as revealed by in situ hybridization. Journal of molecular endocrinology. 2001;26(3):165-174.
  73. Saunders PT, Fisher JS, Sharpe RM, Millar MR. Expression of oestrogen receptor beta (ER beta) occurs in multiple cell types, including some germ cells, in the rat testis. The Journal of endocrinology. 1998;156(3):R13-17.
  74. Pelletier G, El-Alfy M. Immunocytochemical localization of estrogen receptors alpha and beta in the human reproductive organs. The Journal of clinical endocrinology and metabolism. 2000;85(12):4835-4840.
  75. Dumasia K, Kumar A, Deshpande S, Balasinor NH. Estrogen signaling, through estrogen receptor β, regulates DNA methylation and its machinery in male germ line in adult rats. Epigenetics. 2017;12(6):476-483.
  76. Raut S, Kumar AV, Khambata K, Deshpande S, Balasinor NH. Genome-wide identification of estrogen receptor binding sites reveals novel estrogen-responsive pathways in adult male germ cells. Biochem J.2020;477(12):2115-2131.
  77. Chimento A, Sirianni R, Casaburi I, Pezzi V. GPER Signaling in Spermatogenesis and Testicular Tumors. Frontiers in endocrinology. 2014;5:30.
  78. Vaucher L, Funaro MG, Mehta A, Mielnik A, Bolyakov A, Prossnitz ER, Schlegel PN, Paduch DA. Activation of GPER-1 estradiol receptor downregulates production of testosterone in isolated rat Leydig cells and adult human testis. PloS one. 2014;9(4):e92425.
  79. Lucas TF, Pimenta MT, Pisolato R, Lazari MF, Porto CS. 17beta-estradiol signaling and regulation of Sertoli cell function. Spermatogenesis. 2011;1(4):318-324.
  80. Lucas TF, Royer C, Siu ER, Lazari MF, Porto CS. Expression and signaling of G protein-coupled estrogen receptor 1 (GPER) in rat sertoli cells. Biology of reproduction. 2010;83(2):307-317.
  81. Sirianni R, Chimento A, Ruggiero C, De Luca A, Lappano R, Ando S, Maggiolini M, Pezzi V. The novel estrogen receptor, G protein-coupled receptor 30, mediates the proliferative effects induced by 17beta-estradiol on mouse spermatogonial GC-1 cell line. Endocrinology. 2008;149(10):5043-5051.
  82. Chimento A, Sirianni R, Zolea F, Bois C, Delalande C, Ando S, Maggiolini M, Aquila S, Carreau S, Pezzi V. Gper and ESRs are expressed in rat round spermatids and mediate oestrogen-dependent rapid pathways modulating expression of cyclin B1 and Bax. International journal of andrology. 2011;34(5 Pt 1):420-429.
  83. Levallet J, Bilinska B, Mittre H, Genissel C, Fresnel J, Carreau S. Expression and immunolocalization of functional cytochrome P450 aromatase in mature rat testicular cells. Biology of reproduction. 1998;58(4):919-926.
  84. Nitta H, Bunick D, Hess RA, Janulis L, Newton SC, Millette CF, Osawa Y, Shizuta Y, Toda K, Bahr JM. Germ cells of the mouse testis express P450 aromatase. Endocrinology. 1993;132(3):1396-1401.
  85. Cavaco JE, Laurentino SS, Barros A, Sousa M, Socorro S. Estrogen receptors alpha and beta in human testis: both isoforms are expressed. Systems biology in reproductive medicine. 2009;55(4):137-144.
  86. Cunha GR, Li Y, Mei C, Derpinghaus A, Baskin LS. Ontogeny of estrogen receptors in human male and female fetal reproductive tracts. Differentiation. 2021;118:107-131.
  87. Boukari K, Ciampi ML, Guiochon-Mantel A, Young J, Lombes M, Meduri G. Human fetal testis: source of estrogen and target of estrogen action. Human reproduction (Oxford, England). 2007;22(7):1885-1892.
  88. Hess RA, Sharpe RM, Hinton BT. Estrogens and development of the rete testis, efferent ductules, epididymis and vas deferens. Differentiation. 2021;118:41-71.
  89. Berensztein EB, Baquedano MS, Gonzalez CR, Saraco NI, Rodriguez J, Ponzio R, Rivarola MA, Belgorosky A. Expression of aromatase, estrogen receptor alpha and beta, androgen receptor, and cytochrome P-450scc in the human early prepubertal testis. Pediatric research. 2006;60(6):740-744.
  90. Kolasa A, Wiszniewska B, Marchlewicz M, Wenda-Rozewicka L. Localisation of oestrogen receptors (ERalpha and ERbeta) in the human and rat epididymides. Folia morphologica. 2003;62(4):467-469.
  91. Lambard S, Galeraud-Denis I, Saunders PT, Carreau S. Human immature germ cells and ejaculated spermatozoa contain aromatase and oestrogen receptors. Journal of molecular endocrinology. 2004;32(1):279-289.
  92. Aquila S, Sisci D, Gentile M, Middea E, Catalano S, Carpino A, Rago V, Ando S. Estrogen receptor (ER)alpha and ER beta are both expressed in human ejaculated spermatozoa: evidence of their direct interaction with phosphatidylinositol-3-OH kinase/Akt pathway. The Journal of clinical endocrinology and metabolism.2004;89(3):1443-1451.
  93. Aquila S, De Amicis F. Steroid receptors and their ligands: effects on male gamete functions. Experimental cell research. 2014;328(2):303-313.
  94. Solakidi S, Psarra AM, Nikolaropoulos S, Sekeris CE. Estrogen receptors alpha and beta (ERalpha and ERbeta) and androgen receptor (AR) in human sperm: localization of ERbeta and AR in mitochondria of the midpiece. Human reproduction (Oxford, England). 2005;20(12):3481-3487.
  95. Saunders PT, Millar MR, Macpherson S, Irvine DS, Groome NP, Evans LR, Sharpe RM, Scobie GA. ERbeta1 and the ERbeta2 splice variant (ERbetacx/beta2) are expressed in distinct cell populations in the adult human testis. The Journal of clinical endocrinology and metabolism. 2002;87(6):2706-2715.
  96. Aschim EL, Saether T, Wiger R, Grotmol T, Haugen TB. Differential distribution of splice variants of estrogen receptor beta in human testicular cells suggests specific functions in spermatogenesis. The Journal of steroid biochemistry and molecular biology. 2004;92(1-2):97-106.
  97. Ogawa S, Inoue S, Watanabe T, Orimo A, Hosoi T, Ouchi Y, Muramatsu M. Molecular cloning and characterization of human estrogen receptor betacx: a potential inhibitor ofestrogen action in human. Nucleic acids research. 1998;26(15):3505-3512.
  98. Oliveira PF, Alves MG, Martins AD, Correia S, Bernardino RL, Silva J, Barros A, Sousa M, Cavaco JE, Socorro S. Expression pattern of G protein-coupled receptor 30 in human seminiferous tubular cells. General and comparative endocrinology. 2014;201:16-20.
  99. Rago V, Giordano F, Brunelli E, Zito D, Aquila S, Carpino A. Identification of G protein-coupled estrogen receptor in human and pig spermatozoa. Journal of anatomy. 2014;224(6):732-736.
  100. Chimento A, De Luca A, Nocito MC, Avena P, La Padula D, Zavaglia L, Pezzi V. Role of GPER-Mediated Signaling in Testicular Functions and Tumorigenesis. Cells. 2020;9(9).
  101. Carreau S. Germ cells: a new source of estrogens in the male gonad. Molecular and cellular endocrinology.2001;178(1-2):65-72.
  102. Carreau S, Bourguiba S, Lambard S, Galeraud-Denis I, Genissel C, Bilinska B, Benahmed M, Levallet J. Aromatase expression in male germ cells. The Journal of steroid biochemistry and molecular biology.2001;79(1-5):203-208.
  103. Lambard S, Galeraud-Denis I, Bouraima H, Bourguiba S, Chocat A, Carreau S. Expression of aromatase in human ejaculated spermatozoa: a putative marker of motility. Molecular human reproduction. 2003;9(3):117-124.
  104. Meeker JD, Singh NP, Hauser R. Serum concentrations of estradiol and free T4 are inversely correlated with sperm DNA damage in men from an infertility clinic. Journal of andrology. 2008;29(4):379-388.
  105. Pentikainen V, Erkkila K, Suomalainen L, Parvinen M, Dunkel L. Estradiol acts as a germ cell survival factor in the human testis in vitro. The Journal of clinical endocrinology and metabolism. 2000;85(5):2057-2067.
  106. Carreau S, Bouraima-Lelong H, Delalande C. Estrogens: new players in spermatogenesis. Reproductive biology. 2011;11(3):174-193.
  107. Aschim EL, Giwercman A, Stahl O, Eberhard J, Cwikiel M, Nordenskjold A, Haugen TB, Grotmol T, Giwercman YL. The RsaI polymorphism in the estrogen receptor-beta gene is associated with male infertility. The Journal of clinical endocrinology and metabolism. 2005;90(9):5343-5348.
  108. Guarducci E, Nuti F, Becherini L, Rotondi M, Balercia G, Forti G, Krausz C. Estrogen receptor alpha promoter polymorphism: stronger estrogen action is coupled with lower sperm count. Human reproduction (Oxford, England). 2006;21(4):994-1001.
  109. Korach KS. Insights from the study of animals lacking functional estrogen receptor. Science (New York, NY).1994;266(5190):1524-1527.
  110. Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies O. Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proceedings of the National Academy of Sciences of the United States of America. 1993;90(23):11162-11166.
  111. Eddy EM, Washburn TF, Bunch DO, Goulding EH, Gladen BC, Lubahn DB, Korach KS. Targeted disruption of the estrogen receptor gene in male mice causes alteration of spermatogenesis and infertility. Endocrinology.1996;137(11):4796-4805.
  112. Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson JA, Smithies O. Generation and reproductive phenotypes of mice lacking estrogen receptor beta. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(26):15677-15682.
  113. Sharma G, Prossnitz ER. GPER/GPR30 Knockout Mice: Effects of GPER on Metabolism. Methods in molecular biology (Clifton, NJ). 2016;1366:489-502.
  114. Otto C, Fuchs I, Kauselmann G, Kern H, Zevnik B, Andreasen P, Schwarz G, Altmann H, Klewer M, Schoor M, Vonk R, Fritzemeier KH. GPR30 does not mediate estrogenic responses in reproductive organs in mice. Biology of reproduction. 2009;80(1):34-41.
  115. Sharma G, Hu C, Brigman JL, Zhu G, Hathaway HJ, Prossnitz ER. GPER deficiency in male mice results in insulin resistance, dyslipidemia, and a proinflammatory state. Endocrinology. 2013;154(11):4136-4145.
  116. Robertson KM, O'Donnell L, Jones ME, Meachem SJ, Boon WC, Fisher CR, Graves KH, McLachlan RI, Simpson ER. Impairment of spermatogenesis in mice lacking a functional aromatase (cyp 19) gene. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(14):7986-7991.
  117. Murata Y, Robertson KM, Jones ME, Simpson ER. Effect of estrogen deficiency in the male: the ArKO mouse model. Molecular and cellular endocrinology. 2002;193(1-2):7-12.
  118. Sinkevicius KW, Laine M, Lotan TL, Woloszyn K, Richburg JH, Greene GL. Estrogen-dependent and -independent estrogen receptor-alpha signaling separately regulate male fertility. Endocrinology.2009;150(6):2898-2905.
  119. Yao G, Hu S, Yu L, Ru Y, Chen CD, Liu Q, Zhang Y. Genome-Wide Mapping of In Vivo ERα-Binding Sites in Male Mouse Efferent Ductules. Endocrinology. 2017;158(11):3724-3737.
  120. Chimento A, Sirianni R, Casaburi I, Pezzi V. Role of estrogen receptors and g protein-coupled estrogen receptor in regulation of hypothalamus-pituitary-testis axis and spermatogenesis. Frontiers in endocrinology.2014;5:1.
  121. Grumbach MM, Auchus RJ. Estrogen: consequences and implications of human mutations in synthesis and action. The Journal of clinical endocrinology and metabolism. 1999;84(12):4677-4694.
  122. Fisher CR, Graves KH, Parlow AF, Simpson ER. Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(12):6965-6970.
  123. Donaldson KM, Tong SY, Washburn T, Lubahn DB, Eddy EM, Hutson JM, Korach KS. Morphometric study of the gubernaculum in male estrogen receptor mutant mice. Journal of andrology. 1996;17(2):91-95.
  124. Zhou Q, Clarke L, Nie R, Carnes K, Lai LW, Lien YH, Verkman A, Lubahn D, Fisher JS, Katzenellenbogen BS, Hess RA. Estrogen action and male fertility: roles of the sodium/hydrogen exchanger-3 and fluid reabsorption in reproductive tract function. Proceedings of the National Academy of Sciences of the United States of America.2001;98(24):14132-14137.
  125. Lee KH, Finnigan-Bunick C, Bahr J, Bunick D. Estrogen regulation of ion transporter messenger RNA levels in mouse efferent ductules are mediated differentially through estrogen receptor (ER) alpha and ER beta. Biology of reproduction. 2001;65(5):1534-1541.
  126. Lee KH, Hess RA, Bahr JM, Lubahn DB, Taylor J, Bunick D. Estrogen receptor alpha has a functional role in the mouse rete testis and efferent ductules. Biology of reproduction. 2000;63(6):1873-1880.
  127. Mahato D, Goulding EH, Korach KS, Eddy EM. Spermatogenic cells do not require estrogen receptor-alpha for development or function. Endocrinology. 2000;141(3):1273-1276.
  128. Kumar A, Dumasia K, Deshpande S, Raut S, Balasinor NH. Delineating the regulation of estrogen and androgen receptor expression by sex steroids during rat spermatogenesis. The Journal of steroid biochemistry and molecular biology. 2018;182:127-136.
  129. Carreau S, Hess RA. Oestrogens and spermatogenesis. Philosophical transactions of the Royal Society of London Series B, Biological sciences. 2010;365(1546):1517-1535.
  130. Rochira VS, D; Carani, C. Pathophysiology of estrogen action in men. In: Behre ENHM, ed. Testosterone Action, Deficiency, Substitution. Fourth Edition ed. Cambridge, UK: Cambridge University Press; 2012.
  131. Rochira V, Carani C. Estrogen Deficiency in Men. In: Simoni M, Huhtaniemi IT, eds. Endocrinology of the Testis and Male Reproduction. Cham: Springer International Publishing; 2017:797-828.
  132. Rochira V, Madeo B. Estrogens and Male Osteoporosis. In: Ferlin A, Migliaccio S, eds. Male Osteoporosis: Gender Differences in Pathophysiology, Clinical Aspects, Diagnosis and Treatment. Cham: Springer International Publishing; 2020:67-84.
  133. Bernard V, Kherra S, Francou B, Fagart J, Viengchareun S, Guéchot J, Ladjouze A, Guiochon-Mantel A, Korach KS, Binart N, Lombès M, Christin-Maitre S. Familial Multiplicity of Estrogen Insensitivity Associated With a Loss-of-Function ESR1 Mutation. The Journal of clinical endocrinology and metabolism. 2017;102(1):93-99.
  134. Smith EP, Specker B, Bachrach BE, Kimbro KS, Li XJ, Young MF, Fedarko NS, Abuzzahab MJ, Frank GR, Cohen RM, Lubahn DB, Korach KS. Impact on bone of an estrogen receptor-alpha gene loss of function mutation. The Journal of clinical endocrinology and metabolism. 2008;93(8):3088-3096.
  135. Rochira V, Carani C. Aromatase deficiency in men: a clinical perspective. Nature reviews Endocrinology.2009;5(10):559-568.
  136. Bilezikian JP, Morishima A, Bell J, Grumbach MM. Increased bone mass as a result of estrogen therapy in a man with aromatase deficiency. The New England journal of medicine. 1998;339(9):599-603.
  137. Deladoey J, Fluck C, Bex M, Yoshimura N, Harada N, Mullis PE. Aromatase deficiency caused by a novel P450arom gene mutation: impact of absent estrogen production on serum gonadotropin concentration in a boy. The Journal of clinical endocrinology and metabolism. 1999;84(11):4050-4054.
  138. Herrmann BL, Saller B, Janssen OE, Gocke P, Bockisch A, Sperling H, Mann K, Broecker M. Impact of estrogen replacement therapy in a male with congenital aromatase deficiency caused by a novel mutation in the CYP19 gene. The Journal of clinical endocrinology and metabolism. 2002;87(12):5476-5484.
  139. Pura MM, H; Carreau, S; Kottler, ML Clinical findings in an adult man with a novel mutation in the aromatase gene. Paper presented at: Abstract Book of the 85th Annual Meeting of the Endocrine Society2003; Philadelphia, PA, USA
  140. Mittre Herve MH, Kottler ML, Pura M. Human gene mutations. Gene symbol: CYP19. Disease: Aromatase deficiency. Human genetics. 2004;114(2):224.
  141. Bouillon R, Bex M, Vanderschueren D, Boonen S. Estrogens are essential for male pubertal periosteal bone expansion. The Journal of clinical endocrinology and metabolism. 2004;89(12):6025-6029.
  142. Herrmann BL, Janssen OE, Hahn S, Broecker-Preuss M, Mann K. Effects of estrogen replacement therapy on bone and glucose metabolism in a male with congenital aromatase deficiency. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. 2005;37(3):178-183.
  143. Baykan EK, Erdogan M, Ozen S, Darcan S, Saygili LF. Aromatase deficiency, a rare syndrome: case report. Journal of clinical research in pediatric endocrinology. 2013;5(2):129-132.
  144. Pignatti EU, Kursad; Kartal, Ermine; Ajlouni, Kamel; Khawaja, Nahla;Carani, Cesare;  Marino, Marco; Simoni, Manuela; Vighi, Eleonora;  Rochira, Vincenzo. Complete aromatase deficiency in four adult men: detection of a novel mutation and two known mutations in the CYP19A1 gene. Endocrine Abstracts. 2013;32(32):P640.
  145. Bouchoucha N, Samara-Boustani D, Pandey AV, Bony-Trifunovic H, Hofer G, Aigrain Y, Polak M, Flück CE. Characterization of a novel CYP19A1 (aromatase) R192H mutation causing virilization of a 46,XX newborn, undervirilization of the 46,XY brother, but no virilization of the mother during pregnancies. Molecular and cellular endocrinology. 2014;390(1-2):8-17.
  146. Chen Z, Wang O, Nie M, Elison K, Zhou D, Li M, Jiang Y, Xia W, Meng X, Chen S, Xing X. Aromatase deficiency in a Chinese adult man caused by novel compound heterozygous CYP19A1 mutations: effects of estrogen replacement therapy on the bone, lipid, liver and glucose metabolism. Molecular and cellular endocrinology. 2015;399:32-42.
  147. Miedlich SU, Karamooz N, Hammes SR. Aromatase deficiency in a male patient - Case report and review of the literature. Bone. 2016;93:181-186.
  148. Abur U, Atmaca A, Scott H, Gagliardi L, Altundağ E, Akar OS, Bayrak IK, Oğur G. A Male Case of Aromatase Deficiency with a Novel CYP19A1 Mutation. Journal of clinical research in pediatric endocrinology. 2017;9:2-2.
  149. Kim SY, Colaiacovo S, Dave S, Coughlin K, Langdon K, Stein R, Saleh M. Aromatase deficiency in an Ontario Old Order Mennonite family. J Pediatr Endocrinol Metab. 2021;34(12):1615-1618.
  150. Maffei L, Murata Y, Rochira V, Tubert G, Aranda C, Vazquez M, Clyne CD, Davis S, Simpson ER, Carani C. Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment. The Journal of clinical endocrinology and metabolism. 2004;89(1):61-70.
  151. Maffei L, Rochira V, Zirilli L, Antunez P, Aranda C, Fabre B, Simone ML, Pignatti E, Simpson ER, Houssami S, Clyne CD, Carani C. A novel compound heterozygous mutation of the aromatase gene in an adult man: reinforced evidence on the relationship between congenital oestrogen deficiency, adiposity and the metabolic syndrome. Clinical endocrinology. 2007;67(2):218-224.
  152. Lanfranco F, Zirilli L, Baldi M, Pignatti E, Corneli G, Ghigo E, Aimaretti G, Carani C, Rochira V. A novel mutation in the human aromatase gene: insights on the relationship among serum estradiol, longitudinal growth and bone mineral density in an adult man under estrogen replacement treatment. Bone. 2008;43(3):628-635.
  153. Carani C, Rochira V, Faustini-Fustini M, Balestrieri A, Granata AR. Role of oestrogen in male sexual behaviour: insights from the natural model of aromatase deficiency. Clinical endocrinology. 1999;51(4):517-524.
  154. Rochira V, Faustini-Fustini M, Balestrieri A, Carani C. Estrogen replacement therapy in a man with congenital aromatase deficiency: effects of different doses of transdermal estradiol on bone mineral density and hormonal parameters. The Journal of clinical endocrinology and metabolism. 2000;85(5):1841-1845.
  155. Rochira V, Balestrieri A, Faustini-Fustini M, Borgato S, Beck-Peccoz P, Carani C. Pituitary function in a man with congenital aromatase deficiency: effect of different doses of transdermal E2 on basal and stimulated pituitary hormones. The Journal of clinical endocrinology and metabolism. 2002;87(6):2857-2862.
  156. Carani C, Granata AR, Rochira V, Caffagni G, Aranda C, Antunez P, Maffei LE. Sex steroids and sexual desire in a man with a novel mutation of aromatase gene and hypogonadism. Psychoneuroendocrinology.2005;30(5):413-417.
  157. Rochira V, Zirilli L, Genazzani AD, Balestrieri A, Aranda C, Fabre B, Antunez P, Diazzi C, Carani C, Maffei L. Hypothalamic-pituitary-gonadal axis in two men with aromatase deficiency: evidence that circulating estrogens are required at the hypothalamic level for the integrity of gonadotropin negative feedback. European journal of endocrinology / European Federation of Endocrine Societies. 2006;155(4):513-522.
  158. Rochira V, Madeo B, Zirilli L, Caffagni G, Maffei L, Carani C. Oestradiol replacement treatment and glucose homeostasis in two men with congenital aromatase deficiency: evidence for a role of oestradiol and sex steroids imbalance on insulin sensitivity in men. Diabetic medicine : a journal of the British Diabetic Association.2007;24(12):1491-1495.
  159. Rochira V, Zirilli L, Madeo B, Aranda C, Caffagni G, Fabre B, Montangero VE, Roldan EJ, Maffei L, Carani C. Skeletal effects of long-term estrogen and testosterone replacement treatment in a man with congenital aromatase deficiency: evidences of a priming effect of estrogen for sex steroids action on bone. Bone.2007;40(6):1662-1668.
  160. Zirilli L, Maffei L, Meunier PJ, Chavassieux P, Carani C, Rochira V. The effects of long-term raloxifene and estradiol treatments on bone in a patient with congenital aromatase deficiency. Bone. 2009;45(5):827-832.
  161. Rochira V, Balestrieri A, Madeo B, Spaggiari A, Carani C. Congenital estrogen deficiency in men: a new syndrome with different phenotypes; clinical and therapeutic implications in men. Molecular and cellular endocrinology. 2002;193(1-2):19-28.
  162. Fukami M, Ogata T. Congenital disorders of estrogen biosynthesis and action. Best practice & research Clinical endocrinology & metabolism. 2021:101580.
  163. Parween S, Fernández-Cancio M, Benito-Sanz S, Camats N, Rojas Velazquez MN, López-Siguero JP, Udhane SS, Kagawa N, Flück CE, Audí L, Pandey AV. Molecular Basis of CYP19A1 Deficiency in a 46,XX Patient With R550W Mutation in POR: Expanding the PORD Phenotype. The Journal of clinical endocrinology and metabolism. 2020;105(4).
  164. Cooke PS, Walker WH. Nonclassical androgen and estrogen signaling is essential for normal spermatogenesis. Semin Cell Dev Biol. 2021.
  165. Suzuki Y, Sasagawa I, Itoh K, Ashida J, Muroya K, Ogata T. Estrogen receptor alpha gene polymorphism is associated with idiopathic azoospermia. Fertility and sterility. 2002;78(6):1341-1343.
  166. Kukuvitis A, Georgiou I, Bouba I, Tsirka A, Giannouli CH, Yapijakis C, Tarlatzis B, Bontis J, Lolis D, Sofikitis N, Papadimas J. Association of oestrogen receptor alpha polymorphisms and androgen receptor CAG trinucleotide repeats with male infertility: a study in 109 Greek infertile men. International journal of andrology.2002;25(3):149-152.
  167. Lazaros LA, Xita NV, Kaponis AI, Zikopoulos KA, Plachouras NI, Georgiou IA. Estrogen receptor alpha and beta polymorphisms are associated with semen quality. Journal of andrology. 2010;31(3):291-298.
  168. Lazaros L, Xita N, Kaponis A, Hatzi E, Plachouras N, Sofikitis N, Zikopoulos K, Georgiou I. The association of aromatase (CYP19) gene variants with sperm concentration and motility. Asian journal of andrology.2011;13(2):292-297.
  169. Mobasseri N, Nikzad H, Karimian M. Protective effect of oestrogen receptor α-PvuII transition against idiopathic male infertility: a case-control study and meta-analysis. Reprod Biomed Online. 2019;38(4):588-598.
  170. Lee IW, Kuo PH, Su MT, Kuan LC, Hsu CC, Kuo PL. Quantitative trait analysis suggests polymorphisms of estrogen-related genes regulate human sperm concentrations and motility. Human reproduction (Oxford, England). 2011;26(6):1585-1596.
  171. Gonzalez-Unzaga M, Tellez J, Calzada L. Clinical significance of nuclear matrix-estradiol receptor complex in human sperm. Archives of andrology. 2003;49(1):77-81.
  172. Raman JD, Schlegel PN. Aromatase inhibitors for male infertility. The Journal of urology. 2002;167(2 Pt 1):624-629.
  173. Saylam B, Efesoy O, Cayan S. The effect of aromatase inhibitor letrozole on body mass index, serum hormones, and sperm parameters in infertile men. Fertility and sterility. 2011;95(2):809-811.
  174. Shiraishi K, Oka S, Matsuyama H. Testicular Testosterone and Estradiol Concentrations and Aromatase Expression in Men with Nonobstructive Azoospermia. The Journal of clinical endocrinology and metabolism.2021;106(4):e1803-e1815.
  175. Hagiuda J, Ishikawa H, Marumo K. Serum oestradiol levels in male partners of infertile couples. Andrologia.2015;47(6):669-673.
  176. Toppari J, Larsen JC, Christiansen P, Giwercman A, Grandjean P, Guillette LJ, Jr., Jegou B, Jensen TK, Jouannet P, Keiding N, Leffers H, McLachlan JA, Meyer O, Muller J, Rajpert-De Meyts E, Scheike T, Sharpe R, Sumpter J, Skakkebaek NE. Male reproductive health and environmental xenoestrogens. Environmental health perspectives. 1996;104 Suppl 4:741-803.
  177. Pflieger-Bruss S, Schuppe HC, Schill WB. The male reproductive system and its susceptibility to endocrine disrupting chemicals. Andrologia. 2004;36(6):337-345.
  178. Sharpe RM. Do males rely on female hormones? Nature. 1997;390(6659):447-448.
  179. Hrabovszky E, Kalló I, Szlávik N, Keller E, Merchenthaler I, Liposits Z. Gonadotropin-releasing hormone neurons express estrogen receptor-beta. The Journal of clinical endocrinology and metabolism.2007;92(7):2827-2830.
  180. Wolfe A, Wu S. Estrogen receptor-β in the gonadotropin-releasing hormone neuron. Seminars in reproductive medicine. 2012;30(1):23-31.
  181. Mittelman-Smith MA, Williams H, Krajewski-Hall SJ, Lai J, Ciofi P, McMullen NT, Rance NE. Arcuate kisspeptin/neurokinin B/dynorphin (KNDy) neurons mediate the estrogen suppression of gonadotropin secretion and body weight. Endocrinology. 2012;153(6):2800-2812.
  182. Shupnik MA. Oestrogen receptors, receptor variants and oestrogen actions in the hypothalamic-pituitary axis. Journal of neuroendocrinology. 2002;14(2):85-94.
  183. Idan A, Griffiths KA, Harwood DT, Seibel MJ, Turner L, Conway AJ, Handelsman DJ. Long-term effects of dihydrotestosterone treatment on prostate growth in healthy, middle-aged men without prostate disease: a randomized, placebo-controlled trial. Annals of internal medicine. 2010;153(10):621-632.
  184. Finkelstein JS, O'Dea LS, Whitcomb RW, Crowley WF, Jr. Sex steroid control of gonadotropin secretion in the human male. II. Effects of estradiol administration in normal and gonadotropin-releasing hormone-deficient men. The Journal of clinical endocrinology and metabolism. 1991;73(3):621-628.
  185. Finkelstein JS, Whitcomb RW, O'Dea LS, Longcope C, Schoenfeld DA, Crowley WF, Jr. Sex steroid control of gonadotropin secretion in the human male. I. Effects of testosterone administration in normal and gonadotropin-releasing hormone-deficient men. The Journal of clinical endocrinology and metabolism. 1991;73(3):609-620.
  186. Guercio G, Saraco N, Costanzo M, Marino R, Ramirez P, Berensztein E, Rivarola MA, Belgorosky A. Estrogens in Human Male Gonadotropin Secretion and Testicular Physiology From Infancy to Late Puberty. Frontiers in endocrinology. 2020;11:72.
  187. Raven G, de Jong FH, Kaufman JM, de Ronde W. In men, peripheral estradiol levels directly reflect the action of estrogens at the hypothalamo-pituitary level to inhibit gonadotropin secretion. The Journal of clinical endocrinology and metabolism. 2006;91(9):3324-3328.
  188. Bagatell CJ, Dahl KD, Bremner WJ. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. Journal of andrology. 1994;15(1):15-21.
  189. Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley WF, Jr. Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. The Journal of clinical endocrinology and metabolism.2000;85(9):3027-3035.
  190. Pitteloud N, Dwyer AA, DeCruz S, Lee H, Boepple PA, Crowley WF, Jr., Hayes FJ. Inhibition of luteinizing hormone secretion by testosterone in men requires aromatization for its pituitary but not its hypothalamic effects: evidence from the tandem study of normal and gonadotropin-releasing hormone-deficient men. The Journal of clinical endocrinology and metabolism. 2008;93(3):784-791.
  191. Pitteloud N, Dwyer AA, DeCruz S, Lee H, Boepple PA, Crowley WF, Jr., Hayes FJ. The relative role of gonadal sex steroids and gonadotropin-releasing hormone pulse frequency in the regulation of follicle-stimulating hormone secretion in men. The Journal of clinical endocrinology and metabolism. 2008;93(7):2686-2692.
  192. Mauras N, O'Brien KO, Klein KO, Hayes V. Estrogen suppression in males: metabolic effects. The Journal of clinical endocrinology and metabolism. 2000;85(7):2370-2377.
  193. Wickman S, Dunkel L. Inhibition of P450 aromatase enhances gonadotropin secretion in early and midpubertal boys: evidence for a pituitary site of action of endogenous E. The Journal of clinical endocrinology and metabolism. 2001;86(10):4887-4894.
  194. Taxel P, Kennedy DG, Fall PM, Willard AK, Clive JM, Raisz LG. The effect of aromatase inhibition on sex steroids, gonadotropins, and markers of bone turnover in older men. The Journal of clinical endocrinology and metabolism. 2001;86(6):2869-2874.
  195. de Ronde W, de Jong FH. Aromatase inhibitors in men: effects and therapeutic options. Reproductive biology and endocrinology : RB&E. 2011;9:93.
  196. de Ronde W. Therapeutic uses of aromatase inhibitors in men. Current opinion in endocrinology, diabetes, and obesity. 2007;14(3):235-240.
  197. Burnett-Bowie SA, Roupenian KC, Dere ME, Lee H, Leder BZ. Effects of aromatase inhibition in hypogonadal older men: a randomized, double-blind, placebo-controlled trial. Clinical endocrinology. 2009;70(1):116-123.
  198. Leder BZ, Rohrer JL, Rubin SD, Gallo J, Longcope C. Effects of aromatase inhibition in elderly men with low or borderline-low serum testosterone levels. The Journal of clinical endocrinology and metabolism.2004;89(3):1174-1180.
  199. Finkelstein JS, Lee H, Burnett-Bowie SA, Pallais JC, Yu EW, Borges LF, Jones BF, Barry CV, Wulczyn KE, Thomas BJ, Leder BZ. Gonadal steroids and body composition, strength, and sexual function in men. The New England journal of medicine. 2013;369(11):1011-1022.
  200. Awouters M, Vanderschueren D, Antonio L. Aromatase inhibitors and selective estrogen receptor modulators: Unconventional therapies for functional hypogonadism? Andrology. 2020;8(6):1590-1597.
  201. Ide V, Vanderschueren D, Antonio L. Treatment of Men with Central Hypogonadism: Alternatives for Testosterone Replacement Therapy. Int J Mol Sci. 2020;22(1).
  202. McArdle CA, Schomerus E, Groner I, Poch A. Estradiol regulates gonadotropin-releasing hormone receptor number, growth and inositol phosphate production in alpha T3-1 cells. Molecular and cellular endocrinology.1992;87(1-3):95-103.
  203. Hu L, Gustofson RL, Feng H, Leung PK, Mores N, Krsmanovic LZ, Catt KJ. Converse regulatory functions of estrogen receptor-alpha and -beta subtypes expressed in hypothalamic gonadotropin-releasing hormone neurons. Molecular endocrinology (Baltimore, Md). 2008;22(10):2250-2259.
  204. Zhang C, Bosch MA, Levine JE, Ronnekleiv OK, Kelly MJ. Gonadotropin-releasing hormone neurons express K(ATP) channels that are regulated by estrogen and responsive to glucose and metabolic inhibition. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2007;27(38):10153-10164.
  205. Vanderschueren D, Bouillon R. Estrogen deficiency in men is a challenge for both the hypothalamus and pituitary. The Journal of clinical endocrinology and metabolism. 2000;85(9):3024-3026.
  206. Marino R, Perez Garrido N, Costanzo M, Guercio G, Juanes M, Rocco C, Ramirez P, Warman DM, Ciaccio M, Pena G, Feyling JG, Miras M, Rivarola MA, Belgorosky A, Saraco N. Five new cases of 46,XX aromatase deficiency: clinical follow-up from birth to puberty, a novel mutation, and a founder effect. The Journal of clinical endocrinology and metabolism. 2015;100(2):E301-307.
  207. Mongan NP, Tadokoro-Cuccaro R, Bunch T, Hughes IA. Androgen insensitivity syndrome. Best practice & research Clinical endocrinology & metabolism. 2015;29(4):569-580.
  208. Harkonen PL, Makela SI. Role of estrogens in development of prostate cancer. The Journal of steroid biochemistry and molecular biology. 2004;92(4):297-305.
  209. Kowalska K, Piastowska-Ciesielska AW. Oestrogens and oestrogen receptors in prostate cancer. SpringerPlus.2016;5:522.
  210. McPherson SJ, Wang H, Jones ME, Pedersen J, Iismaa TP, Wreford N, Simpson ER, Risbridger GP. Elevated androgens and prolactin in aromatase-deficient mice cause enlargement, but not malignancy, of the prostate gland. Endocrinology. 2001;142(6):2458-2467.
  211. Li X, Nokkala E, Yan W, Streng T, Saarinen N, Warri A, Huhtaniemi I, Santti R, Makela S, Poutanen M. Altered structure and function of reproductive organs in transgenic male mice overexpressing human aromatase. Endocrinology. 2001;142(6):2435-2442.
  212. McPherson SJ, Ellem SJ, Patchev V, Fritzemeier KH, Risbridger GP. The role of Eralpha and ERbeta in the prostate: insights from genetic models and isoform-selective ligands. Ernst Schering Foundation symposium proceedings. 2006(1):131-147.
  213. Bosland MC. The role of steroid hormones in prostate carcinogenesis. Journal of the National Cancer Institute Monographs. 2000(27):39-66.
  214. Ellem SJ, Schmitt JF, Pedersen JS, Frydenberg M, Risbridger GP. Local aromatase expression in human prostate is altered in malignancy. The Journal of clinical endocrinology and metabolism. 2004;89(5):2434-2441.
  215. Kanda S, Tsuchiya N, Narita S, Inoue T, Huang M, Chiba S, Akihama S, Saito M, Numakura K, Tsuruta H, Satoh S, Saito S, Ohyama C, Arai Y, Ogawa O, Habuchi T. Effects of functional genetic polymorphisms in the CYP19A1 gene on prostate cancer risk and survival. Int J Cancer. 2015;136(1):74-82.
  216. Beuten J, Gelfond JA, Franke JL, Weldon KS, Crandall AC, Johnson-Pais TL, Thompson IM, Leach RJ. Single and multigenic analysis of the association between variants in 12 steroid hormone metabolism genes and risk of prostate cancer. Cancer Epidemiol Biomarkers Prev. 2009;18(6):1869-1880.
  217. Bosland MC, Ford H, Horton L. Induction at high incidence of ductal prostate adenocarcinomas in NBL/Cr and Sprague-Dawley Hsd:SD rats treated with a combination of testosterone and estradiol-17 beta or diethylstilbestrol. Carcinogenesis. 1995;16(6):1311-1317.
  218. Qu LG, Wardan H, Davis ID, Pezaro C, Sluka P. Effects of estrogen receptor signaling on prostate cancer carcinogenesis. Transl Res. 2020;222:56-66.
  219. Lau KM, To KF. Importance of Estrogenic Signaling and Its Mediated Receptors in Prostate Cancer. Int J Mol Sci. 2016;17(9).
  220. Nakajima Y, Osakabe A, Waku T, Suzuki T, Akaogi K, Fujimura T, Homma Y, Inoue S, Yanagisawa J. Estrogen Exhibits a Biphasic Effect on Prostate Tumor Growth through the Estrogen Receptor β-KLF5 Pathway. Mol Cell Biol. 2016;36(1):144-156.
  221. Dias JP, Melvin D, Shardell M, Ferrucci L, Chia CW, Gharib M, Egan JM, Basaria S. Effects of Transdermal Testosterone Gel or an Aromatase Inhibitor on Prostate Volume in Older Men. The Journal of clinical endocrinology and metabolism. 2016;101(4):1865-1871.
  222. Xu D, Wu Y, Shen H, Qian S, Qi J. High serum concentration of estradiol may be a risk factor of prostate enlargement in aging male in China. Aging Male. 2020;23(1):1-6.
  223. Reis LO, Zani EL, García-Perdomo HA. Estrogen therapy in patients with prostate cancer: a contemporary systematic review. Int Urol Nephrol. 2018;50(6):993-1003.
  224. Langley RE, Gilbert DC, Duong T, Clarke NW, Nankivell M, Rosen SD, Mangar S, Macnair A, Sundaram SK, Laniado ME, Dixit S, Madaan S, Manetta C, Pope A, Scrase CD, McKay S, Muazzam IA, Collins GN, Worlding J, Williams ST, Paez E, Robinson A, McFarlane J, Deighan JV, Marshall J, Forcat S, Weiss M, Kockelbergh R, Alhasso A, Kynaston H, Parmar M. Transdermal oestradiol for androgen suppression in prostate cancer: long-term cardiovascular outcomes from the randomised Prostate Adenocarcinoma Transcutaneous Hormone (PATCH) trial programme. Lancet (London, England). 2021;397(10274):581-591.
  225. Brodie AM, Son C, King DA, Meyer KM, Inkster SE. Lack of evidence for aromatase in human prostatic tissues: effects of 4-hydroxyandrostenedione and other inhibitors on androgen metabolism. Cancer research.1989;49(23):6551-6555.
  226. Rochira V, Zirilli L, Madeo B, Balestrieri A, Granata AR, Carani C. Sex steroids and sexual desire mechanism. Journal of endocrinological investigation. 2003;26(3 Suppl):29-36.
  227. Robbins A. Androgens and male sexual behavior from mice to men. Trends in endocrinology and metabolism: TEM. 1996;7(9):345-350.
  228. van Anders SM. Beyond masculinity: testosterone, gender/sex, and human social behavior in a comparative context. Frontiers in neuroendocrinology. 2013;34(3):198-210.
  229. Funk CH, B; Lejwa, A. The male hormone. American Journal of Physiology. 1930;92:440-449.
  230. Granata AR, Rochira V, Lerchl A, Marrama P, Carani C. Relationship between sleep-related erections and testosterone levels in men. Journal of andrology. 1997;18(5):522-527.
  231. Rochira V, Balestrieri A, Madeo B, Granata AR, Carani C. Sildenafil improves sleep-related erections in hypogonadal men: evidence from a randomized, placebo-controlled, crossover study of a synergic role for both testosterone and sildenafil on penile erections. Journal of andrology. 2006;27(2):165-175.
  232. Gooren LJ. Clinical practice. Care of transsexual persons. The New England journal of medicine.2011;364(13):1251-1257.
  233. Van Goozen SH, Cohen-Kettenis PT, Gooren LJ, Frijda NH, Van de Poll NE. Gender differences in behaviour: activating effects of cross-sex hormones. Psychoneuroendocrinology. 1995;20(4):343-363.
  234. Gooren LJ, Giltay EJ. Men and women, so different, so similar: observations from cross-sex hormone treatment of transsexual subjects. Andrologia. 2014;46(5):570-575.
  235. Ramasamy R, Scovell JM, Kovac JR, Lipshultz LI. Elevated serum estradiol is associated with higher libido in men on testosterone supplementation therapy. European urology. 2014;65(6):1224-1225.
  236. Cunningham GR, Stephens-Shields AJ, Rosen RC, Wang C, Ellenberg SS, Matsumoto AM, Bhasin S, Molitch ME, Farrar JT, Cella D, Barrett-Connor E, Cauley JA, Cifelli D, Crandall JP, Ensrud KE, Fluharty L, Gill TM, Lewis CE, Pahor M, Resnick SM, Storer TW, Swerdloff RS, Anton S, Basaria S, Diem S, Tabatabaie V, Hou X, Snyder PJ. Association of sex hormones with sexual function, vitality, and physical function of symptomatic older men with low testosterone levels at baseline in the testosterone trials. The Journal of clinical endocrinology and metabolism. 2015;100(3):1146-1155.
  237. Sartorius GA, Ly LP, Handelsman DJ. Male sexual function can be maintained without aromatization: randomized placebo-controlled trial of dihydrotestosterone (DHT) in healthy, older men for 24 months. The journal of sexual medicine. 2014;11(10):2562-2570.
  238. O'Connor DB, Lee DM, Corona G, Forti G, Tajar A, O'Neill TW, Pendleton N, Bartfai G, Boonen S, Casanueva FF, Finn JD, Giwercman A, Han TS, Huhtaniemi IT, Kula K, Labrie F, Lean ME, Punab M, Silman AJ, Vanderschueren D, Wu FC. The relationships between sex hormones and sexual function in middle-aged and older European men. The Journal of clinical endocrinology and metabolism. 2011;96(10):E1577-1587.
  239. Meyer-Bahlburg HF. Sex steroids and variants of gender identity. Endocrinology and metabolism clinics of North America. 2013;42(3):435-452.
  240. Bao AM, Swaab DF. Sexual differentiation of the human brain: relation to gender identity, sexual orientation and neuropsychiatric disorders. Frontiers in neuroendocrinology. 2011;32(2):214-226.
  241. Gorski R. Sexual differentiation of the endocrine brain and its control. In: Motta M, ed. Brain Endocrinology. 2nd ed. New York: Raven Press; 1991:71-104.
  242. Pilgrim C, Reisert I. Differences between male and female brains--developmental mechanisms and implications. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme.1992;24(8):353-359.
  243. Garcia-Falgueras A, Swaab DF. Sexual hormones and the brain: an essential alliance for sexual identity and sexual orientation. Endocrine development. 2010;17:22-35.
  244. Dorner G. Neuroendocrine response to estrogen and brain differentiation in heterosexuals, homosexuals, and transsexuals. Archives of sexual behavior. 1988;17(1):57-75.
  245. Roselli CE, Liu M, Hurn PD. Brain aromatization: classic roles and new perspectives. Seminars in reproductive medicine. 2009;27(3):207-217.
  246. Swaab DF, Wolff SEC, Bao AM. Sexual differentiation of the human hypothalamus: Relationship to gender identity and sexual orientation. Handb Clin Neurol. 2021;181:427-443.
  247. Roselli CE, Estill CT, Stadelman HL, Meaker M, Stormshak F. Separate critical periods exist for testosterone-induced differentiation of the brain and genitals in sheep. Endocrinology. 2011;152(6):2409-2415.
  248. LeVay S. A difference in hypothalamic structure between heterosexual and homosexual men. Science (New York, NY). 1991;253(5023):1034-1037.
  249. Wu MV, Manoli DS, Fraser EJ, Coats JK, Tollkuhn J, Honda S, Harada N, Shah NM. Estrogen masculinizes neural pathways and sex-specific behaviors. Cell. 2009;139(1):61-72.
  250. Roselli CE, Larkin K, Resko JA, Stellflug JN, Stormshak F. The volume of a sexually dimorphic nucleus in the ovine medial preoptic area/anterior hypothalamus varies with sexual partner preference. Endocrinology.2004;145(2):478-483.
  251. Troisi R, Palmer JR, Hatch EE, Strohsnitter WC, Huo D, Hyer M, Fredriksen-Goldsen KI, Hoover R, Titus L. Gender Identity and Sexual Orientation Identity in Women and Men Prenatally Exposed to Diethylstilbestrol. Archives of sexual behavior. 2020;49(2):447-454.
  252. Wright CL, Schwarz JS, Dean SL, McCarthy MM. Cellular mechanisms of estradiol-mediated sexual differentiation of the brain. Trends in endocrinology and metabolism: TEM. 2010;21(9):553-561.
  253. Wallen K, Hassett JM. Sexual differentiation of behaviour in monkeys: role of prenatal hormones. Journal of neuroendocrinology. 2009;21(4):421-426.
  254. Jannini EA, Blanchard R, Camperio-Ciani A, Bancroft J. Male homosexuality: nature or culture? The journal of sexual medicine. 2010;7(10):3245-3253.
  255. Carani CR, V; Granata, ARM. Sexuality and erectile dysfunction. In: Wass JS, SM ed. Oxford Textbook of Endocrinology and Diabetes. 2nd ed. Oxford, UK: Oxford University Press; 2011:1449-1458.
  256. Corona G, Isidori AM, Buvat J, Aversa A, Rastrelli G, Hackett G, Rochira V, Sforza A, Lenzi A, Mannucci E, Maggi M. Testosterone supplementation and sexual function: a meta-analysis study. The journal of sexual medicine. 2014;11(6):1577-1592.
  257. Corona G, Isidori AM, Aversa A, Burnett AL, Maggi M. Endocrinologic Control of Men's Sexual Desire and Arousal/Erection. The journal of sexual medicine. 2016;13(3):317-337.
  258. Buvat J, Maggi M, Gooren L, Guay AT, Kaufman J, Morgentaler A, Schulman C, Tan HM, Torres LO, Yassin A, Zitzmann M. Endocrine aspects of male sexual dysfunctions. The journal of sexual medicine. 2010;7(4 Pt 2):1627-1656.
  259. Honda S, Harada N, Ito S, Takagi Y, Maeda S. Disruption of sexual behavior in male aromatase-deficient mice lacking exons 1 and 2 of the cyp19 gene. Biochemical and biophysical research communications.1998;252(2):445-449.
  260. Ogawa S, Chester AE, Hewitt SC, Walker VR, Gustafsson JA, Smithies O, Korach KS, Pfaff DW. Abolition of male sexual behaviors in mice lacking estrogen receptors alpha and beta (alpha beta ERKO). Proceedings of the National Academy of Sciences of the United States of America. 2000;97(26):14737-14741.
  261. Luisi M, Franchi F. Double-blind group comparative study of testosterone undecanoate and mesterolone in hypogonadal male patients. Journal of endocrinological investigation. 1980;3(3):305-308.
  262. Davidson JM, Camargo C, Smith ER, Kwan M. Maintenance of sexual function in a castrated man treated with ovarian steroids. Archives of sexual behavior. 1983;12(3):263-274.
  263. Cunningham GR, Stephens-Shields AJ, Rosen RC, Wang C, Bhasin S, Matsumoto AM, Parsons JK, Gill TM, Molitch ME, Farrar JT, Cella D, Barrett-Connor E, Cauley JA, Cifelli D, Crandall JP, Ensrud KE, Gallagher L, Zeldow B, Lewis CE, Pahor M, Swerdloff RS, Hou X, Anton S, Basaria S, Diem SJ, Tabatabaie V, Ellenberg SS, Snyder PJ. Testosterone Treatment and Sexual Function in Older Men With Low Testosterone Levels. The Journal of clinical endocrinology and metabolism. 2016;101(8):3096-3104.
  264. Wibowo E, Schellhammer P, Wassersug RJ. Role of estrogen in normal male function: clinical implications for patients with prostate cancer on androgen deprivation therapy. The Journal of urology. 2011;185(1):17-23.
  265. Hsu B, Cumming RG, Blyth FM, Naganathan V, Le Couteur DG, Seibel MJ, Waite LM, Handelsman DJ. The longitudinal relationship of sexual function and androgen status in older men: the Concord Health and Ageing in Men Project. The Journal of clinical endocrinology and metabolism. 2015;100(4):1350-1358.
  266. Wu F, Chen T, Mao S, Jiang H, Ding Q, Xu G. Levels of estradiol and testosterone are altered in Chinese men with sexual dysfunction. Andrology. 2016;4(5):932-938.
  267. Chen T, Wu F, Wang X, Ma G, Xuan X, Tang R, Ding S, Lu J. Different levels of estradiol are correlated with sexual dysfunction in adult men. Sci Rep. 2020;10(1):12660.
  268. Chen HR, Tian RH, Li P, Chen HX, Xia SJ, Li Z. Estradiol is an independent risk factor for organic erectile dysfunction in eugonadal young men. Asian journal of andrology. 2020;22(6):636-641.
  269. El-Sakka AI. Impact of the association between elevated oestradiol and low testosterone levels on erectile dysfunction severity. Asian journal of andrology. 2013;15(4):492-496.
  270. Vignozzi L, Filippi S, Comeglio P, Cellai I, Morelli A, Marchetta M, Maggi M. Estrogen mediates metabolic syndrome-induced erectile dysfunction: a study in the rabbit. The journal of sexual medicine. 2014;11(12):2890-2902.
  271. Castelló-Porcar AM, Martínez-Jabaloyas JM. Testosterone/estradiol ratio, is it useful in the diagnosis of erectile dysfunction and low sexual desire? Aging Male. 2016;19(4):254-258.
  272. Crescioli C, Maggi M, Vannelli GB, Ferruzzi P, Granchi S, Mancina R, Muratori M, Forti G, Serio M, Luconi M. Expression of functional estrogen receptors in human fetal male external genitalia. The Journal of clinical endocrinology and metabolism. 2003;88(4):1815-1824.
  273. Dietrich W, Haitel A, Huber JC, Reiter WJ. Expression of estrogen receptors in human corpus cavernosum and male urethra. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.2004;52(3):355-360.
  274. Goyal HO, Braden TD, Williams CS, Williams JW. Role of estrogen in induction of penile dysmorphogenesis: a review. Reproduction (Cambridge, England). 2007;134(2):199-208.
  275. Mowa CN, Jesmin S, Miyauchi T. The penis: a new target and source of estrogen in male reproduction. Histology and histopathology. 2006;21(1):53-67.
  276. Mauvais-Jarvis F. Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity. Trends in endocrinology and metabolism: TEM. 2011;22(1):24-33.
  277. Gibb FW, Homer NZ, Faqehi AM, Upreti R, Livingstone DE, McInnes KJ, Andrew R, Walker BR. Aromatase Inhibition Reduces Insulin Sensitivity in Healthy Men. The Journal of clinical endocrinology and metabolism.2016;101(5):2040-2046.
  278. Dias JP, Shardell MD, Carlson OD, Melvin D, Caturegli G, Ferrucci L, Chia CW, Egan JM, Basaria S. Testosterone vs. aromatase inhibitor in older men with low testosterone: effects on cardiometabolic parameters. Andrology. 2017;5(1):31-40.
  279. Juang PS, Peng S, Allehmazedeh K, Shah A, Coviello AD, Herbst KL. Testosterone with dutasteride, but not anastrazole, improves insulin sensitivity in young obese men: a randomized controlled trial. The journal of sexual medicine. 2014;11(2):563-573.
  280. Dhindsa S, Furlanetto R, Vora M, Ghanim H, Chaudhuri A, Dandona P. Low estradiol concentrations in men with subnormal testosterone concentrations and type 2 diabetes. Diabetes Care. 2011;34(8):1854-1859.
  281. Chan YX, Knuiman MW, Hung J, Divitini ML, Handelsman DJ, Beilby JP, McQuillan B, Yeap BB. Testosterone, dihydrotestosterone and estradiol are differentially associated with carotid intima-media thickness and the presence of carotid plaque in men with and without coronary artery disease. Endocr J. 2015;62(9):777-786.
  282. van Koeverden ID, de Bakker M, Haitjema S, van der Laan SW, de Vries JPM, Hoefer IE, de Borst GJ, Pasterkamp G, den Ruijter HM. Testosterone to oestradiol ratio reflects systemic and plaque inflammation and predicts future cardiovascular events in men with severe atherosclerosis. Cardiovasc Res. 2019;115(2):453-462.
  283. Figtree GA, Ngo DTM, Bubb KJ. Testosterone to estradiol ratio and plaque inflammation: mechanistic insights and biomarker potential? Cardiovasc Res. 2019;115(2):255-257.
  284. Travison TG, O'Donnell CJ, Bhasin S, Massaro JM, Hoffmann U, Vasan RS, D'Agostino RB, Sr., Basaria S. Circulating Sex Steroids and Vascular Calcification in Community-Dwelling Men: The Framingham Heart Study. The Journal of clinical endocrinology and metabolism. 2016;101(5):2160-2167.
  285. Taylor AP, Lee H, Webb ML, Joffe H, Finkelstein JS. Effects of Testosterone and Estradiol Deficiency on Vasomotor Symptoms in Hypogonadal Men. The Journal of clinical endocrinology and metabolism.2016;101(9):3479-3486.
  286. Rochira V, Kara E, Carani C. The endocrine role of estrogens on human male skeleton. International journal of endocrinology. 2015;2015:165215.
  287. Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Borjesson AE, Ohlsson C. Sex steroid actions in male bone. Endocrine reviews. 2014;35(6):906-960.
  288. Khosla S. Estrogen and bone: insights from estrogen-resistant, aromatase-deficient, and normal men. Bone.2008;43(3):414-417.
  289. Khosla S. Update on estrogens and the skeleton. The Journal of clinical endocrinology and metabolism.2010;95(8):3569-3577.
  290. Rochira V, Balestrieri A, Madeo B, Zirilli L, Granata AR, Carani C. Osteoporosis and male age-related hypogonadism: role of sex steroids on bone (patho)physiology. European journal of endocrinology / European Federation of Endocrine Societies. 2006;154(2):175-185.
  291. Rochira V, Antonio L, Vanderschueren D. EAA clinical guideline on management of bone health in the andrological outpatient clinic. Andrology. 2018;6(2):272-285.
  292. Rochira V. Late-onset Hypogonadism: Bone health. Andrology. 2020;8(6):1539-1550.
  293. Finkelstein JS, Lee H, Leder BZ, Burnett-Bowie SA, Goldstein DW, Hahn CW, Hirsch SC, Linker A, Perros N, Servais AB, Taylor AP, Webb ML, Youngner JM, Yu EW. Gonadal steroid-dependent effects on bone turnover and bone mineral density in men. The Journal of clinical investigation. 2016;126(3):1114-1125.
  294. Decaroli MC, Rochira V. Aging and sex hormones in males. Virulence. 2017;8(5):545-570.
  295. Russell N, Hoermann R, Cheung AS, Ching M, Zajac JD, Handelsman DJ, Grossmann M. Short-term effects of transdermal estradiol in men undergoing androgen deprivation therapy for prostate cancer: a randomized placebo-controlled trial. European journal of endocrinology / European Federation of Endocrine Societies.2018;178(5):565-576.
  296. Khalil R, Antonio L, Laurent MR, David K, Kim NR, Evenepoel P, Eisenhauer A, Heuser A, Cavalier E, Khosla S, Claessens F, Vanderschueren D, Decallonne B. Early effects of androgen deprivation on bone and mineral homeostasis in adult men: a prospective cohort study. European journal of endocrinology / European Federation of Endocrine Societies. 2020;183(2):181-189.
  297. Decaroli MC, De Vincentis S, Rochira V. Aging and sex hormones in males. Vitam Horm. 2021;115:333-366.
  298. Langley RE, Kynaston HG, Alhasso AA, Duong T, Paez EM, Jovic G, Scrase CD, Robertson A, Cafferty F, Welland A, Carpenter R, Honeyfield L, Abel RL, Stone M, Parmar MK, Abel PD. A Randomised Comparison Evaluating Changes in Bone Mineral Density in Advanced Prostate Cancer: Luteinising Hormone-releasing Hormone Agonists Versus Transdermal Oestradiol. European urology. 2016;69(6):1016-1025.
  299. Handelsman DJ, Yeap B, Flicker L, Martin S, Wittert GA, Ly LP. Age-specific population centiles for androgen status in men. European journal of endocrinology / European Federation of Endocrine Societies.2015;173(6):809-817.
  300. Frederiksen H, Johannsen TH, Andersen SE, Albrethsen J, Landersoe SK, Petersen JH, Andersen AN, Vestergaard ET, Schorring ME, Linneberg A, Main KM, Andersson AM, Juul A. Sex-specific Estrogen Levels and Reference Intervals from Infancy to Late Adulthood Determined by LC-MS/MS. The Journal of clinical endocrinology and metabolism. 2020;105(3):754-768.
  301. Vottero A, Rochira V, Capelletti M, Viani I, Zirilli L, Neri TM, Carani C, Bernasconi S, Ghizzoni L. Aromatase is differentially expressed in peripheral blood leukocytes from children, and adult female and male subjects. European journal of endocrinology / European Federation of Endocrine Societies. 2006;154(3):425-431.
  302. Ongphiphadhanakul B, Rajatanavin R, Chanprasertyothin S, Piaseu N, Chailurkit L. Serum oestradiol and oestrogen-receptor gene polymorphism are associated with bone mineral density independently of serum testosterone in normal males. Clinical endocrinology. 1998;49(6):803-809.
  303. Khosla S, Riggs BL, Atkinson EJ, Oberg AL, Mavilia C, Del Monte F, Melton LJ, 3rd, Brandi ML. Relationship of estrogen receptor genotypes to bone mineral density and to rates of bone loss in men. The Journal of clinical endocrinology and metabolism. 2004;89(4):1808-1816.
  304. Gennari L, Masi L, Merlotti D, Picariello L, Falchetti A, Tanini A, Mavilia C, Del Monte F, Gonnelli S, Lucani B, Gennari C, Brandi ML. A polymorphic CYP19 TTTA repeat influences aromatase activity and estrogen levels in elderly men: effects on bone metabolism. The Journal of clinical endocrinology and metabolism.2004;89(6):2803-2810.
  305. Van Pottelbergh I, Goemaere S, Kaufman JM. Bioavailable estradiol and an aromatase gene polymorphism are determinants of bone mineral density changes in men over 70 years of age. The Journal of clinical endocrinology and metabolism. 2003;88(7):3075-3081.
  306. Eriksson AL, Perry JRB, Coviello AD, Delgado GE, Ferrucci L, Hoffman AR, Huhtaniemi IT, Ikram MA, Karlsson MK, Kleber ME, Laughlin GA, Liu Y, Lorentzon M, Lunetta KL, Mellstrom D, Murabito JM, Murray A, Nethander M, Nielson CM, Prokopenko I, Pye SR, Raffel LJ, Rivadeneira F, Srikanth P, Stolk L, Teumer A, Travison TG, Uitterlinden AG, Vaidya D, Vanderschueren D, Zmuda JM, Marz W, Orwoll ES, Ouyang P, Vandenput L, Wu FCW, de Jong FH, Bhasin S, Kiel DP, Ohlsson C. Genetic Determinants of Circulating Estrogen Levels and Evidence of a Causal Effect of Estradiol on Bone Density in Men. The Journal of clinical endocrinology and metabolism. 2018;103(3):991-1004.
  307. Schmitz D, Ek WE, Berggren E, Höglund J, Karlsson T, Johansson Å. Genome-wide Association Study of Estradiol Levels and the Causal Effect of Estradiol on Bone Mineral Density. The Journal of clinical endocrinology and metabolism. 2021;106(11):e4471-e4486.
  308. Nethander M, Vandenput L, Eriksson AL, Windahl S, Funck-Brentano T, Ohlsson C. Evidence of a Causal Effect of Estradiol on Fracture Risk in Men. The Journal of clinical endocrinology and metabolism. 2019;104(2):433-442.
  309. Van Caenegem E, Wierckx K, Taes Y, Schreiner T, Vandewalle S, Toye K, Kaufman JM, T'Sjoen G. Preservation of volumetric bone density and geometry in trans women during cross-sex hormonal therapy: a prospective observational study. Osteoporos Int. 2015;26(1):35-47.
  310. Laurent MR, Gielen E, Vanderschueren D. Estrogens, the be-all and end-all of male hypogonadal bone loss? Osteoporos Int. 2015;26(1):29-33.
  311. Björntorp P. The regulation of adipose tissue distribution in humans. Int J Obes Relat Metab Disord.1996;20(4):291-302.
  312. Isidori AM, Giannetta E, Greco EA, Gianfrilli D, Bonifacio V, Isidori A, Lenzi A, Fabbri A. Effects of testosterone on body composition, bone metabolism and serum lipid profile in middle-aged men: a meta-analysis. Clinical endocrinology. 2005;63(3):280-293.
  313. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(23):12729-12734.
  314. Misso ML, Murata Y, Boon WC, Jones ME, Britt KL, Simpson ER. Cellular and molecular characterization of the adipose phenotype of the aromatase-deficient mouse. Endocrinology. 2003;144(4):1474-1480.
  315. Chao J, Rubinow KB, Kratz M, Amory JK, Matsumoto AM, Page ST. Short-Term Estrogen Withdrawal Increases Adiposity in Healthy Men. The Journal of clinical endocrinology and metabolism. 2016;101(10):3724-3731.
  316. Smith MR. Changes in body composition during hormonal therapy for prostate cancer. Clin Prostate Cancer.2003;2(1):18-21.
  317. Smith MR. Changes in fat and lean body mass during androgen-deprivation therapy for prostate cancer. Urology. 2004;63(4):742-745.
  318. Haseen F, Murray LJ, Cardwell CR, O'Sullivan JM, Cantwell MM. The effect of androgen deprivation therapy on body composition in men with prostate cancer: systematic review and meta-analysis. J Cancer Surviv.2010;4(2):128-139.
  319. Russell N, Hoermann R, Cheung AS, Zajac JD, Handelsman DJ, Grossmann M. Effects of estradiol on fat in men undergoing androgen deprivation therapy: a randomized trial. European journal of endocrinology / European Federation of Endocrine Societies. 2021;186(1):9-23.
  320. Gava G, Mancini I, Orsili I, Bertelloni S, Alvisi S, Seracchioli R, Meriggiola MC. Bone mineral density, body composition and metabolic profiles in adult women with complete androgen insensitivity syndrome and removed gonads using oral or transdermal estrogens. European journal of endocrinology / European Federation of Endocrine Societies. 2019;181(6):711-718.
  321. Birzniece V, Ho KKY. Sex steroids and the GH axis: Implications for the management of hypopituitarism. Best practice & research Clinical endocrinology & metabolism. 2017;31(1):59-69.
  322. Nilsson M, Dahlman I, Rydén M, Nordström EA, Gustafsson JA, Arner P, Dahlman-Wright K. Oestrogen receptor alpha gene expression levels are reduced in obese compared to normal weight females. Int J Obes (Lond). 2007;31(6):900-907.
  323. Drew BG, Hamidi H, Zhou Z, Villanueva CJ, Krum SA, Calkin AC, Parks BW, Ribas V, Kalajian NY, Phun J, Daraei P, Christofk HR, Hewitt SC, Korach KS, Tontonoz P, Lusis AJ, Slamon DJ, Hurvitz SA, Hevener AL. Estrogen receptor (ER)α-regulated lipocalin 2 expression in adipose tissue links obesity with breast cancer progression. J Biol Chem. 2015;290(9):5566-5581.
  324. Zhou Z, Moore TM, Drew BG, Ribas V, Wanagat J, Civelek M, Segawa M, Wolf DM, Norheim F, Seldin MM, Strumwasser AR, Whitney KA, Lester E, Reddish BR, Vergnes L, Reue K, Rajbhandari P, Tontonoz P, Lee J, Mahata SK, Hewitt SC, Shirihai O, Gastonbury C, Small KS, Laakso M, Jensen J, Lee S, Drevon CA, Korach KS, Lusis AJ, Hevener AL. Estrogen receptor α controls metabolism in white and brown adipocytes by regulating Polg1 and mitochondrial remodeling. Sci Transl Med. 2020;12(555).
  325. Fuller-Jackson JP, Dordevic AL, Clarke IJ, Henry BA. Effect of sex and sex steroids on brown adipose tissue heat production in humans. European journal of endocrinology / European Federation of Endocrine Societies.2020;183(3):343-355.
  326. López M, Tena-Sempere M. Estrogens and the control of energy homeostasis: a brain perspective. Trends in endocrinology and metabolism: TEM. 2015;26(8):411-421.
  327. Davis KE, M DN, Sun K, W MS, J DB, J AZ, Zeve D, L DH, D WC, L MG, Xu Y, Z VW, S AK, Clegg DJ. The sexually dimorphic role of adipose and adipocyte estrogen receptors in modulating adipose tissue expansion, inflammation, and fibrosis. Mol Metab. 2013;2(3):227-242.
  328. Sample CH, Davidson TL. Considering sex differences in the cognitive controls of feeding. Physiol Behav.2018;187:97-107.
  329. Rivera HM, Stincic TL. Estradiol and the control of feeding behavior. Steroids. 2018;133:44-52.
  330. LaBar KS, Gitelman DR, Parrish TB, Kim YH, Nobre AC, Mesulam MM. Hunger selectively modulates corticolimbic activation to food stimuli in humans. Behav Neurosci. 2001;115(2):493-500.
  331. Biegon A, Alia-Klein N, Alexoff DL, Fowler JS, Kim SW, Logan J, Pareto D, Preston-Campbell R, Wang GJ, Hildebrandt T. Relationship of estrogen synthesis capacity in the brain with obesity and self-control in men and women. Proc Natl Acad Sci U S A. 2020;117(37):22962-22966.
  332. Hampton T. How Targeting Fat Cells' Estrogen Receptors Could Fight Obesity. Jama. 2020;324(21):2146.
  333. Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci.2020;21(10):535-550.
  334. Yeap BB, Hui J, Knuiman MW, Handelsman DJ, Flicker L, Divitini ML, Arscott GM, McLennan SV, Twigg SM, Almeida OP, Hankey GJ, Golledge J, Norman PE, Beilby JP. Cross-sectional associations of sex hormones with leucocyte telomere length, a marker of biological age, in a community-based cohort of older men. Clinical endocrinology. 2019;90(4):562-569.
  335. Yeap BB, Hui J, Knuiman MW, C SAP, Ken KYH, Flicker L, Divitini ML, Arscott GM, Twigg SM, Almeida OP, Hankey GJ, Golledge J, Beilby JP. Associations of plasma IGF1, IGFBP3 and estradiol with leucocyte telomere length, a marker of biological age, in men. European journal of endocrinology / European Federation of Endocrine Societies. 2020;182(1):23-33.
  336. Titus-Ernstoff L, Troisi R, Hatch EE, Palmer JR, Hyer M, Kaufman R, Adam E, Noller K, Hoover RN. Birth defects in the sons and daughters of women who were exposed in utero to diethylstilbestrol (DES). International journal of andrology. 2010;33(2):377-384.
  337. Streng T, Li X, Lehtoranta M, Makela S, Poutanen M, Talo A, Tekmal RR, Santti R. Infravesical obstruction in aromatase over expressing transgenic male mice with increased ratio of serum estrogen-to-androgen concentration. The Journal of urology. 2002;168(1):298-302.
  338. Kumar V, Misro MM, Datta K. Simultaneous accumulation of hyaluronan binding protein 1 (HABP1/p32/gC1qR) and apoptotic induction of germ cells in cryptorchid testis. Journal of andrology. 2012;33(1):114-121.
  339. Toppari J, Skakkebaek NE. Sexual differentiation and environmental endocrine disrupters. Bailliere's clinical endocrinology and metabolism. 1998;12(1):143-156.
  340. Virtanen HE, Adamsson A. Cryptorchidism and endocrine disrupting chemicals. Molecular and cellular endocrinology. 2012;355(2):208-220.
  341. Toppari J, Rodprasert W, Koskenniemi JJ. Exposure Variation and Endocrine Disruption of the Male Reproductive System. Hormone research in paediatrics. 2016;85(6).
  342. Norgil Damgaard I, Main KM, Toppari J, Skakkebaek NE. Impact of exposure to endocrine disrupters in utero and in childhood on adult reproduction. Best practice & research Clinical endocrinology & metabolism.2002;16(2):289-309.
  343. Martin OV, Shialis T, Lester JN, Scrimshaw MD, Boobis AR, Voulvoulis N. Testicular dysgenesis syndrome and the estrogen hypothesis: a quantitative meta-analysis. Environmental health perspectives. 2008;116(2):149-157.
  344. Den Hond E, Tournaye H, De Sutter P, Ombelet W, Baeyens W, Covaci A, Cox B, Nawrot TS, Van Larebeke N, D'Hooghe T. Human exposure to endocrine disrupting chemicals and fertility: A case-control study in male subfertility patients. Environment international. 2015;84:154-160.
  345. Shozu M, Sebastian S, Takayama K, Hsu WT, Schultz RA, Neely K, Bryant M, Bulun SE. Estrogen excess associated with novel gain-of-function mutations affecting the aromatase gene. The New England journal of medicine. 2003;348(19):1855-1865.
  346. Hemsell DL, Edman CD, Marks JF, Siiteri PK, MacDonald PC. Massive extranglandular aromatization of plasma androstenedione resulting in feminization of a prepubertal boy. The Journal of clinical investigation.1977;60(2):455-464.
  347. Berkovitz GD, Guerami A, Brown TR, MacDonald PC, Migeon CJ. Familial gynecomastia with increased extraglandular aromatization of plasma carbon19-steroids. J Clin Invest. 1985;75(6):1763-1769.
  348. Stratakis CA, Vottero A, Brodie A, Kirschner LS, DeAtkine D, Lu Q, Yue W, Mitsiades CS, Flor AW, Chrousos GP. The aromatase excess syndrome is associated with feminization of both sexes and autosomal dominant transmission of aberrant P450 aromatase gene transcription. The Journal of clinical endocrinology and metabolism. 1998;83(4):1348-1357.
  349. Leiberman E, Zachmann M. Familial adrenal feminization probably due to increased steroid aromatization. Hormone research. 1992;37(3):96-102.
  350. Fukami M, Shozu M, Soneda S, Kato F, Inagaki A, Takagi H, Hanaki K, Kanzaki S, Ohyama K, Sano T, Nishigaki T, Yokoya S, Binder G, Horikawa R, Ogata T. Aromatase excess syndrome: identification of cryptic duplications and deletions leading to gain of function of CYP19A1 and assessment of phenotypic determinants. The Journal of clinical endocrinology and metabolism. 2011;96(6):E1035-1043.
  351. Shihara D, Miyado M, Nakabayashi K, Shozu M, Ogata T, Nagasaki K, Fukami M. Aromatase excess syndrome in a family with upstream deletion of CYP19A1. Clinical endocrinology. 2014;81(2):314-316.
  352. Tan X, Wu X, Chen J, Wu Y, Li S, Chen X, Zhang X. Aromatase excess syndrome in a Chinese boy due to a novel duplication at 15q21.2. J Pediatr Endocrinol Metab. 2019;32(1):85-88.
  353. Binder G, Nakamura A, Schweizer R, Ogata T, Fukami M, Nagasaki K. Long-term Effect of Aromatase Inhibition in Aromatase Excess Syndrome. The Journal of clinical endocrinology and metabolism. 2021;106(5):1491-1500.
  354. Klinefelter HF RE, Albright F. Syndrome characterized by gynecomastia, aspermatogenesis without A-Leydigism, and increased excretion of follicle stimulating hormone. Journal of Clinical Endocrinology.1942;2(11):615-627.
  355. Lanfranco F, Kamischke A, Zitzmann M, Nieschlag E. Klinefelter's syndrome. Lancet (London, England).2004;364(9430):273-283.
  356. Wistuba J. Animal models for Klinefelter's syndrome and their relevance for the clinic. Molecular human reproduction. 2010;16(6):375-385.
  357. Bonomi MR, V; Pasquali, D; Balercia, G; Jannini, EA; Ferlin, A. Klinefelter Syndrome (KS): genetics, clinical phenotype and hypogonadism. Journal of Endocrinological Investigation. 2016.
  358. Santi D, De Vincentis S, Scaltriti S, Rochira V. Relative hyperestrogenism in Klinefelter Syndrome: results from a meta-analysis. Endocrine. 2019;64(2):209-219.
  359. Jiang DD, Swenson E, Mason M, Turner KR, Dugi DD, Hedges JC, Hecht SL. Effects of Estrogen on Spermatogenesis in Transgender Women. Urology. 2019;132:117-122.
  360. Leavy M, Trottmann M, Liedl B, Reese S, Stief C, Freitag B, Baugh J, Spagnoli G, Kölle S. Effects of Elevated β-Estradiol Levels on the Functional Morphology of the Testis - New Insights. Sci Rep. 2017;7:39931.
  361. Matoso A, Khandakar B, Yuan S, Wu T, Wang LJ, Lombardo KA, Mangray S, Mannan A, Yakirevich E. Spectrum of findings in orchiectomy specimens of persons undergoing gender confirmation surgery. Hum Pathol. 2018;76:91-99.
  362. Vereecke G, Defreyne J, Van Saen D, Collet S, Van Dorpe J, T'Sjoen G, Goossens E. Characterisation of testicular function and spermatogenesis in transgender women. Human reproduction (Oxford, England).2021;36(1):5-15.
  363. Schneider F, Kliesch S, Schlatt S, Neuhaus N. Andrology of male-to-female transsexuals: influence of cross-sex hormone therapy on testicular function. Andrology. 2017;5(5):873-880.
  364. Mezzullo M, Pelusi C, Fazzini A, Repaci A, Di Dalmazi G, Gambineri A, Pagotto U, Fanelli F. Female and male serum reference intervals for challenging sex and precursor steroids by liquid chromatography - tandem mass spectrometry. The Journal of steroid biochemistry and molecular biology. 2020;197:105538.
  365. Balestrieri A, Faustini-Fustini M, Rochira V, Carani C. Clinical implications and management of oestrogen deficiency in the male. Clinical endocrinology. 2001;54(4):431-432.
  366. Rosner W, Hankinson SE, Sluss PM, Vesper HW, Wierman ME. Challenges to the measurement of estradiol: an endocrine society position statement. The Journal of clinical endocrinology and metabolism.2013;98(4):1376-1387.
  367. Handelsman DJ, Newman JD, Jimenez M, McLachlan R, Sartorius G, Jones GR. Performance of direct estradiol immunoassays with human male serum samples. Clinical chemistry. 2014;60(3):510-517.
  368. Klein KO, Baron J, Colli MJ, McDonnell DP, Cutler GB, Jr. Estrogen levels in childhood determined by an ultrasensitive recombinant cell bioassay. The Journal of clinical investigation. 1994;94(6):2475-2480.
  369. Huhtaniemi IT, Tajar A, Lee DM, O'Neill TW, Finn JD, Bartfai G, Boonen S, Casanueva FF, Giwercman A, Han TS, Kula K, Labrie F, Lean ME, Pendleton N, Punab M, Silman AJ, Vanderschueren D, Forti G, Wu FC. Comparison of serum testosterone and estradiol measurements in 3174 European men using platform immunoassay and mass spectrometry; relevance for the diagnostics in aging men. European journal of endocrinology / European Federation of Endocrine Societies. 2012;166(6):983-991.
  370. Ketha H, Girtman A, Singh RJ. Estradiol assays--The path ahead. Steroids. 2015;99(Pt A):39-44.
  371. Simoni MF, F; Roli, L; Pagotto, U. Methodology for measuring testosterone, dihydrotestosterone and sex hormone-binding globulin in a clinical setting. In: Behre ENHM, ed. Testosterone Action, Deficiency, Substitution. Fourth Edition ed. Cambridge, UK: Cambridge University Press; 2012:60-86.
  372. Pagotto U, Fanelli F, Granata AR. Hormonal Laboratory Examination. In: Simoni M, Huhtaniemi IT, eds. Endocrinology of the Testis and Male Reproduction. Cham: Springer International Publishing; 2017:495-516.
  373. Aguirre LE, Colleluori G, Fowler KE, Jan IZ, Villareal K, Qualls C, Robbins D, Villareal DT, Armamento-Villareal R. High aromatase activity in hypogonadal men is associated with higher spine bone mineral density, increased truncal fat and reduced lean mass. European journal of endocrinology / European Federation of Endocrine Societies. 2015;173(2):167-174.
  374. Aguirre LE, Colleluori G, Robbins D, Dorin R, Shah VO, Chen R, Jan IZ, Qualls C, Villareal DT, Armamento-Villareal R. Bone and body composition response to testosterone therapy vary according to polymorphisms in the CYP19A1 gene. Endocrine. 2019;65(3):692-706.
  375. Stephens-Shields AJ, Snyder PJ, Ellenberg SS, Taylor L, Bhasin S. Relation of Testosterone, Dihydrotestosterone, and Estradiol With Changes in Outcomes Measures in the Testosterone Trials. The Journal of clinical endocrinology and metabolism. 2022;107(5):1257-1269.
  376. Garg H, Kumar R. Empirical Drug Therapy for Idiopathic Male Infertility: What is the New Evidence? Urology.2015;86(6):1065-1075.
  377. Yang C, Li P, Li Z. Clinical application of aromatase inhibitors to treat male infertility. Hum Reprod Update.2021;28(1):30-50.
  378. Kamischke A, Nieschlag E. Analysis of medical treatment of male infertility. Hum Reprod. 1999;14 Suppl 1:1-23.
  379. Vandekerckhove P, Lilford R, Vail A, Hughes E. Clomiphene or tamoxifen for idiopathic oligo/asthenospermia. The Cochrane database of systematic reviews. 2000(2):CD000151.
  380. Vandekerckhove P, Lilford R, Vail A, Hughes E. WITHDRAWN: Clomiphene or tamoxifen for idiopathic oligo/asthenospermia. The Cochrane database of systematic reviews. 2007(4):CD000151.
  381. Gregoriou O, Bakas P, Grigoriadis C, Creatsa M, Hassiakos D, Creatsas G. Changes in hormonal profile and seminal parameters with use of aromatase inhibitors in management of infertile men with low testosterone to estradiol ratios. Fertility and sterility. 2012;98(1):48-51.
  382. Mellinger RC, Thompson RJ. The effect of clomiphene citrate in male infertility. Fertility and sterility.1966;17(1):94-103.
  383. Kotoulas IG, Cardamakis E, Michopoulos J, Mitropoulos D, Dounis A. Tamoxifen treatment in male infertility. I. Effect on spermatozoa. Fertility and sterility. 1994;61(5):911-914.
  384. Clark RV, Sherins RJ. Treatment of men with idiopathic oligozoospermic infertility using the aromatase inhibitor, testolactone. Results of a double-blinded, randomized, placebo-controlled trial with crossover. Journal of andrology. 1989;10(3):240-247.
  385. Unal D, Yeni E, Verit A, Karatas OF. Clomiphene citrate versus varicocelectomy in treatment of subclinical varicocele: a prospective randomized study. International journal of urology : official journal of the Japanese Urological Association. 2001;8(5):227-230.
  386. Cappon GD, Horimoto M, Hurtt ME. Reproductive toxicity assessment of lasofoxifene, a selective estrogen receptor modulator (SERM), in male rats. Birth defects research Part B, Developmental and reproductive toxicology. 2004;71(3):142-149.
  387. Katz DJ, Nabulsi O, Tal R, Mulhall JP. Outcomes of clomiphene citrate treatment in young hypogonadal men. BJU international. 2012;110(4):573-578.
  388. Ghanem H, Shaeer O, El-Segini A. Combination clomiphene citrate and antioxidant therapy for idiopathic male infertility: a randomized controlled trial. Fertility and sterility. 2010;93(7):2232-2235.
  389. Surbone A, Vaucher L, Primi MP, Leyvraz C, Pitteloud N, Ballabeni P, Mathevet P, Vulliemoz N. Clomiphene citrate effect on testosterone level and semen parameters in 18 infertile men with low testosterone level and normal/low gonadotropines level. Eur J Obstet Gynecol Reprod Biol. 2019;238:104-109.
  390. Sharma D, Zillioux J, Khourdaji I, Reines K, Wheeler K, Costabile R, Kavoussi P, Smith R. Improvements in semen parameters in men treated with clomiphene citrate-A retrospective analysis. Andrologia.2019;51(5):e13257.
  391. Patel DP, Brant WO, Myers JB, Presson AP, Johnstone EB, Dorais JA, Aston KI, Carrell DT, Hotaling JM. The safety and efficacy of clomiphene citrate in hypoandrogenic and subfertile men. Int J Impot Res. 2015;27(6):221-224.
  392. Helo S, Ellen J, Mechlin C, Feustel P, Grossman M, Ditkoff E, McCullough A. A Randomized Prospective Double-Blind Comparison Trial of Clomiphene Citrate and Anastrozole in Raising Testosterone in Hypogonadal Infertile Men. The journal of sexual medicine. 2015;12(8):1761-1769.
  393. Cannarella R, Condorelli RA, Mongioì LM, Barbagallo F, Calogero AE, La Vignera S. Effects of the selective estrogen receptor modulators for the treatment of male infertility: a systematic review and meta-analysis. Expert Opin Pharmacother. 2019;20(12):1517-1525.
  394. Gundewar T, Kuchakulla M, Ramasamy R. A paradoxical decline in semen parameters in men treated with clomiphene citrate: A systematic review. Andrologia. 2021;53(1):e13848.
  395. Wiehle RD, Fontenot GK, Wike J, Hsu K, Nydell J, Lipshultz L. Enclomiphene citrate stimulates testosterone production while preventing oligospermia: a randomized phase II clinical trial comparing topical testosterone. Fertility and sterility. 2014;102(3):720-727.
  396. Earl JA, Kim ED. Enclomiphene citrate: A treatment that maintains fertility in men with secondary hypogonadism. Expert Rev Endocrinol Metab. 2019;14(3):157-165.
  397. Adamopoulos DA, Pappa A, Billa E, Nicopoulou S, Koukkou E, Michopoulos J. Effectiveness of combined tamoxifen citrate and testosterone undecanoate treatment in men with idiopathic oligozoospermia. Fertility and sterility. 2003;80(4):914-920.
  398. Moein MR, Tabibnejad N, Ghasemzadeh J. Beneficial effect of tamoxifen on sperm recovery in infertile men with nonobstructive azoospermia. Andrologia. 2012;44 Suppl 1:194-198.
  399. Boonyarangkul A, Vinayanuvattikhun N, Chiamchanya C, Visutakul P. Comparative Study of the Effects of Tamoxifen Citrate and Folate on Semen Quality of the Infertile Male with Semen Abnormality. Journal of the Medical Association of Thailand = Chotmaihet thangphaet. 2015;98(11):1057-1063.
  400. Tsourdi E, Kourtis A, Farmakiotis D, Katsikis I, Salmas M, Panidis D. The effect of selective estrogen receptor modulator administration on the hypothalamic-pituitary-testicular axis in men with idiopathic oligozoospermia. Fertility and sterility. 2009;91(4 Suppl):1427-1430.
  401. Chua ME, Escusa KG, Luna S, Tapia LC, Dofitas B, Morales M. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology.2013;1(5):749-757.
  402. Cavallini G, Biagiotti G, Bolzon E. Multivariate analysis to predict letrozole efficacy in improving sperm count of non-obstructive azoospermic and cryptozoospermic patients: a pilot study. Asian journal of andrology.2013;15(6):806-811.
  403. Shoshany O, Abhyankar N, Mufarreh N, Daniel G, Niederberger C. Outcomes of anastrozole in oligozoospermic hypoandrogenic subfertile men. Fertility and sterility. 2017;107(3):589-594.
  404. Shuling L, Sie Kuei ML, Saffari SE, Jiayun Z, Yeun TT, Leng JPW, Viardot-Foucault V, Nadarajah S, Chan JKY, Hao TH. Do men with normal testosterone-oestradiol ratios benefit from letrozole for the treatment of male infertility? Reprod Biomed Online. 2019;38(1):39-45.
  405. Peivandi S, Jafarpour H, Abbaspour M, Ebadi A. Effect of letrozole on spermogram parameters and hormonal profile in infertile men: A clinical trial study. Endocr Regul. 2019;53(4):231-236.
  406. Shah T, Nyirenda T, Shin D. Efficacy of anastrozole in the treatment of hypogonadal, subfertile men with body mass index ≥25 kg/m(2). Transl Androl Urol. 2021;10(3):1222-1228.
  407. Kooshesh L, Bahmanpour S, Zeighami S, Nasr-Esfahani MH. Effect of Letrozole on sperm parameters, chromatin status and ROS level in idiopathic Oligo/Astheno/Teratozoospermia. Reproductive biology and endocrinology : RB&E. 2020;18(1):47.
  408. Ribeiro MA, Gameiro LF, Scarano WR, Briton-Jones C, Kapoor A, Rosa MB, El Dib R. Aromatase inhibitors in the treatment of oligozoospermic or azoospermic men: a systematic review of randomized controlled trials. JBRA Assist Reprod. 2016;20(2):82-88.
  409. Kumar R, Gautam G, Gupta NP. Drug therapy for idiopathic male infertility: rationale versus evidence. J Urol.2006;176(4 Pt 1):1307-1312.
  410. Chandrapal JC, Nielson S, Patel DP, Zhang C, Presson AP, Brant WO, Myers JB, Hotaling JM. Characterising the safety of clomiphene citrate in male patients through prostate-specific antigen, haematocrit, and testosterone levels. BJU international. 2016;118(6):994-1000.
  411. Wibowo E, Pollock PA, Hollis N, Wassersug RJ. Tamoxifen in men: a review of adverse events. Andrology.2016;4(5):776-788.
  412. Bradley KL, Tyldesley S, Speers CH, Woods R, Villa D. Contemporary systemic therapy for male breast cancer. Clinical breast cancer. 2014;14(1):31-39.
  413. Loves S, de Jong J, van Sorge A, Telting D, Tack CJ, Hermus A, Westerterp K, de Boer H. Somatic and psychological effects of low-dose aromatase inhibition in men with obesity-related hypogonadotropic hypotestosteronemia. European journal of endocrinology / European Federation of Endocrine Societies.2013;169(5):705-714.
  414. Lapauw B, T'Sjoen G, Mahmoud A, Kaufman JM, Ruige JB. Short-term aromatase inhibition: effects on glucose metabolism and serum leptin levels in young and elderly men. European journal of endocrinology / European Federation of Endocrine Societies. 2009;160(3):397-402.
  415. Hero M, Toiviainen-Salo S, Wickman S, Makitie O, Dunkel L. Vertebral morphology in aromatase inhibitor-treated males with idiopathic short stature or constitutional delay of puberty. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2010;25(7):1536-1543.
  416. Burnett-Bowie SA, McKay EA, Lee H, Leder BZ. Effects of aromatase inhibition on bone mineral density and bone turnover in older men with low testosterone levels. The Journal of clinical endocrinology and metabolism.2009;94(12):4785-4792.
  417. Hero M, Ankarberg-Lindgren C, Taskinen MR, Dunkel L. Blockade of oestrogen biosynthesis in peripubertal boys: effects on lipid metabolism, insulin sensitivity, and body composition. European journal of endocrinology / European Federation of Endocrine Societies. 2006;155(3):453-460.
  418. Allasia S, Motta G, Mirabelli M, Tagliabue MP, Lanfranco F. A case of deep vein thrombosis in a young male treated with tamoxifen for idiopathic infertility. Asian journal of andrology. 2017;19(5):615-616.
  419. Sun D, Jin B, Cai B, Deng W. Deep vein thrombosis in a nonobstructive azoospermia male taking tamoxifen: a rare case report. Transl Androl Urol. 2020;9(4):1769-1772.
  420. Colpi GM, Francavilla S, Haidl G, Link K, Behre HM, Goulis DG, Krausz C, Giwercman A. European Academy of Andrology guideline Management of oligo-astheno-teratozoospermia. Andrology. 2018;6(4):513-524.
  421. Ferlin A, Calogero AE, Krausz C, Lombardo F, Paoli D, Rago R, Scarica C, Simoni M, Foresta C, Rochira V, Sbardella E, Francavilla S, Corona G. Management of male factor infertility: position statement from the Italian Society of Andrology and Sexual Medicine (SIAMS) : Endorsing Organization: Italian Society of Embryology, Reproduction, and Research (SIERR). Journal of endocrinological investigation. 2022;45(5):1085-1113.
  422. Hsu B, Cumming RG, Naganathan V, Blyth FM, Le Couteur DG, Hirani V, Waite LM, Seibel MJ, Handelsman DJ. Temporal Changes in Androgens and Estrogens Are Associated With All-Cause and Cause-Specific Mortality in Older Men. The Journal of clinical endocrinology and metabolism. 2016;101(5):2201-2210.
  423. Loh NY, Humphreys E, Karpe F, Tomlinson JW, Noordam R, Christodoulides C. Sex hormones, adiposity, and metabolic traits in men and women: a Mendelian randomisation study. European journal of endocrinology / European Federation of Endocrine Societies. 2022;186(3):407-416.

Lysosomal Acid Lipase Deficiency

ABSTRACT

 

Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive genetic disease with variable presentation which often leads to severe morbidity and mortality. More than 100 LIPA loss of function mutations have been identified, the most common reported mutation being a splice junction mutation in exon 8. The true prevalence of the disease is unknown, but is estimated to be between 1:40,000 to 1:300,000. Infantile-onset LAL-D is generally fatal within the first 12 months of life. Common presenting symptoms in the late-onset form include dyslipidemia (elevated LDL-C, low HDL-C), elevated liver transaminases, hepatomegaly, and splenomegaly.  Prior to the availability of enzyme-replacement therapy, individuals with LAL-D were treated with lipid lowering medication, liver transplant, and stem cell transplant, none of which corrected the multisystem nature of the disorder. Sebelipase alfa (Kanuma®), a recombinant human lysosomal acid lipase, was approved by the FDA in 2015 to treat LAL-D. Phase 3 studies have shown an improvement in lipid parameters and liver enzymes. Long term studies demonstrating the safety and efficacy of sebelipase alfa in infants, children and adults are ongoing.

 

INTRODUCTION

 

Lysosomal acid lipase deficiency (LAL-D) is a rare, heterogeneous, autosomal recessive genetic disease, the manifestations of which include a clinical continuum. LAL-D is characterized by accumulation of cholesteryl esters and triglycerides primarily in the liver and spleen, but with involvement of other organs as well. Clinically, LAL-D is under-recognized, leading to a delay in diagnosis. It is often mistaken for more common conditions with similar clinical and laboratory findings, such as heterozygous familial hypercholesterolemia (FH) and non-alcoholic fatty liver disease (NAFLD) (1,2). Correct diagnosis and timely intervention are critical to prolonging life and improving outcomes.

 

Similar to other lysosomal storage disorders, LAL-D presents across a clinical spectrum from infancy to adulthood. Historically, affected infants who presented within the first year of life were known as Wolman Disease while those who symptoms were delayed until childhood were referred to as cholesteryl ester storage disease [CESD]. Wolman disease, which has a rapidly progressive course, was first described in 1956. Affected infants have severe malnutrition, adrenal calcifications, hepatosplenomegaly, and death within the first few months of life (3). In contrast, CESD is seen as having a variable clinical spectrum with recognition of the disorder occurring from childhood into adulthood. Fredrickson, Schiff, Langeron, and Infante were the first to describe CESD in individuals with presentation from the first to fourth decades of life, and noted them to be less severe than those described by Wolman (4-6).

 

INHERITANCE AND GENETICS

 

LAL-D is an autosomal recessive disease that arises from mutations at the LAL locus on chromosome 10q23.2.  Affected individuals are either homozygous or compound heterozygous for LIPA mutations, with more than 100 LIPA mutations having been identified (7).

 

Lysosomal acid lipase (LAL) plays a central role in intracellular lipid metabolism (8,9). LAL is the only lipase contained within lysosomes that hydrolyzes cholesteryl esters and triglycerides.  After cleavage by LAL, free cholesterol and fatty acids exit the lysosome to enter the cytosol (Figure 1). These cleaved products play an important role in cholesterol homeostasis. Free cholesterol interacts with transcription factors (sterol regulatory element binding proteins [SREBPs]) to modulate production of intracellular cholesterol. As intracellular free cholesterol increases, there is a down regulation of LDL receptors mediated by SREBP-2, resulting in less LDL entering the cell. Additionally, there is inhibition of hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase, resulting in decreased cholesterol production, as well as stimulation of acyl-cholesterol acyltransferase (leading to increased cholesterol esterification). Finally, increased intracellular fatty acid leads to inhibition of triglyceride and phospholipid production and decreased fatty acid synthesis (10-12).

 

Deficiency of LAL results in diminished or absent hydrolysis of cholesteryl esters and triglycerides, trapping cholesterol esters and TG within the lysosome. This results in a decrease in cytosolic free cholesterol and a compensatory, upregulation in the cholesterol synthetic pathway (HMG CoA reductase activity) and endocytosis via increased LDL receptors. There is increased production of apolipoprotein B and very low-density lipoprotein (VLDL-C) (13-15). The dysregulated expression of the LDL-cholesterol-dependent ATP binding cassette transporter 1 (ABCA1), similar to that seen in Niemann-Pick type C1, results in decreased levels of HDL-C (16). The characteristic dyslipidemia seen in individuals with LAL-D includes elevated total cholesterol, elevated LDL-C, and low HDL-C (2).

 

Figure 1. Cellular Cholesterol Homeostasis in Heathy Individuals and Patients with LAL-D

The true incidence of LAL-D is unknown. Estimates suggest overall disease prevalence between 1:40,000 to 1:300,000, depending on ethnicity and geographical location (1,2,17). The most commonly inherited defect is a splice junction mutation in exon 8, E8SJM (c.894G>A). It is assumed that 50-70% of adults and children with LAL-D have E8SJM (17,18). Studies in the general population have shown that the estimated frequency of E8SJM allele is 0.0013 in Caucasians, 0.0017 in US Hispanics, 0.0010 in US Ashkenazi Jews, and 0.0005 in Asians (19).  Population screening for E8SJM among healthy West German individuals reveal a heterozygote frequency of ~ 1:200 individuals. Jewish infants of Iraqi or Iranian origin appear to be at high risk for LAL-D with an estimated incidence of 1:4,200 in the Los Angeles community (20).

 

A study attempting to identify the prevalence of LAL-D from patients with abnormal results in laboratory databases (elevated LDL-C and abnormalities on liver tests) identified a total of 1825 patients who subsequently underwent a dried blood spot sample for determination of LAL enzyme activity. No cases of LAL-D were identified. The results of this study demonstrate the potential of databases in helping to identify patients with specific patterns of results to allow targeted testing for possible causes of disease. Biochemical screening suggested that the gene frequency of LAL deficiency in adults is less than 1:100 (21). Additionally, histopathology databases of liver biopsies were analyzed searching for patients with features of 'microvesicular cirrhosis' or 'cryptogenic cirrhosis'. DNA was available from six patients and two were homozygous for LAL c.46A>C;p.Thr16Pro, an unclassified variant in exon 2 (21). The results of these studies suggest the potential of databases in helping to identify patients with specific laboratory results or those who had certain biopsy findings to allow targeted testing for possible causes of disease.

 

PRESENTATION

 

The symptoms of LAL-D are quite varied, and are related to the age that clinical manifestations first appear (Figure 2).  Individuals who present within the first few days to first month of life often have vomiting, diarrhea, hepatosplenomegaly, abdominal distention, and severe failure to thrive. The first symptom observed is usually vomiting, which has been described as forceful and persistent. Accompanying these symptoms are usually watery diarrhea and low-grade fever. Symptoms generally persist despite multiple medical interventions and may lead to severe malnutrition. A hallmark of infantile-onset LAL-D is adrenal enlargement and calcification, often seen on imaging, but not required for diagnosis. Calcifications of the adrenal gland as well as adrenal insufficiency have been documented. Few patients survive beyond 12 months of age (2,3,22), with those that have growth failure often dying by four months of age (23).

 

In contrast, the clinical presentation and progression of LAL-D can be variable in older children and adults. However, there are common clinical manifestations that have been reported in this group of patients. In a review of 71 patients two thirds presented with their first symptoms before the age of 5 years. Hepatomegaly was present in all the patients; 86% had splenomegaly.  Gastrointestinal symptoms were present in 30% and included vomiting and diarrhea [18%], failure to thrive [16%], abdominal pain [10%], gastrointestinal bleeding [8%], and gallbladder disease [4%]. Elevation of cholesterol was present in 90% (24). In a separate review of 135 patients, the median age of onset of symptoms was 5 years with a range from birth to 68 years.  Hepatomegaly was present in 99.3% of patients. The most common extrahepatic findings were steatorrhea, poor growth, gallbladder dysfunction, and cardiovascular disease. Total cholesterol was elevated in all 110 patients (1). 

 

The disease severity is likely dependent on the efficiency of alternative pathways, but not on the level of residual enzyme activity (25). In adults, the most frequent symptoms are abdominal pain, hepatomegaly, and laboratory abnormalities that include increased levels of transaminases and cholesterol. Differential diagnostic considerations include autoimmune hepatitis, NASH, alpha1-antitrypsin deficiency, and Wilson disease. Of concern is the potential for premature atherosclerosis in affected individuals.  Although the occurrence of cardiovascular events has not been extensively studied, case reports and observational studies have documented the presence of arterial plaque and atheroma at a very early age (26-28). As a result, many patients with this disease have been prescribed lipid-lowering medications (1). While lipid lowering in the setting of LAL-A has been variable, statins increase hepatic uptake of LDL and, as a consequence, may worsen the lipid overload (29). It is important to note that seven asymptomatic adults, diagnosed in the third to sixth decade of life, have been reported.  All were coincidently found to have confirmed LAL-D, yet none had detectable hepatomegaly (28). 

 

The most consistent biochemical abnormalities seen in late onset LAL-D include elevated liver transaminases and plasma lipids. In a study of 49 patients designed to characterize clinical manifestations of LAL-D, mean ALT, AST, and GGT were 92.4, 87.8, and 52.2 U/L at the first measurement. In this study elevated GGT levels were uncommon (only 20% had values > 40 U/L) (30). In another study, liver dysfunction occurred in 100% of 135 patients and 73% of the 11 reported deaths were due to liver failure (1). Mean LDL-C at the time of first measurement was 202.9 mg/dL, and reported as abnormal in 64.4% of patients. Mean total cholesterol was 269.5 mg/dL and was abnormal in 62.5%. Mean HDL-C was 37.5 mg/dL and abnormal in 43.5% of patients (30). The lipid abnormalities seen most closely resemble type II-b dyslipidemia (31).  Although elevated LDL-C seems to be a feature of LAL-D, it remains unclear whether or not LAL-D is a cause of early atherosclerosis. Case reports and several autopsy studies have noted aortic stenosis and found narrowing of the coronary artery secondary to atheromatous plaque in patients with LAL-D (2,32).

 

Figure 2. Clinical Presentation of LAL-D

 

On gross examination, the liver of patients with LAL-D is enlarged and appears greasy. Liver biopsies in paraffin sections have a predominance of microvesicular steatosis, which is uniform.  Microvesicular steatosis, per se, is not pathognomonic of LAL-D, being found in other liver diseases as well. Foamy macrophages, containing lipid and ceroid, are present in the sinusoids and portal tracts (Figure 3).  Staining for LAMP1, LAMP2, and LIMP2, or with a lysosomal luminal protein (cathepsin D), can assist identifying lipid accumulation as lysosomal, may help differentiate LAL-D from other causes of microvesicular steatosis. Another pathognomonic feature of LAL-D is birefringent cholesterol ester crystals in hepatocytes and Kupffer cells, using polarized light on electron microscopy. The liver disease generally progresses to fibrosis followed by micronodular cirrhosis (1,2,33).

 

Figure 3. Liver Biopsies in Patients with LAL-D. A) Image of the portal tract and hepatocytes with mainly microvesicular steatosis. With microvesicular steatosis, the fat does not cause the nucleus to be pushed out to the side. B) Larger magnification of the portal tract. FM points to the foamy appearing cytoplasm, these are macrophages with something being stored in them. GC is pointing to a giant cell.

 

DIAGNOSTIC TESTS

 

LAL-D can be diagnosed by demonstrating deficient LAL enzyme activity, as well as by genetic testing identifying mutations of the LIPA gene. Historically, enzyme activity was measured in cultured fibroblasts, peripheral leukocytes, or liver tissue. Various lipase substrates, which were not specific for LAL, were used. In the review by Bernstein, enzyme activities were reported in 114 patients and ranged from undetectable to 16% of normal, with values for most patients being between <1%-10%. However, given assay variability, residual enzyme activity is not predictive of disease severity nor can it be compared from one lab to another (1).  

 

A newer method has been developed to determine LAL activity. This method measures LAL activity in dried blood spots (DBS), and uses Lalistat 2, a highly specific inhibitor of LAL. LAL activity is determined by comparing total lipase activity to lipase activity with Lalistat 2. This method is able to differentiate normal from affected individuals. This DBS technique has advantages over the fibroblast/peripheral leukocyte based test including small sample size, the ability to transport the specimen to the testing facility at ambient temperature, and sample stability (34). This blood test is available at a number of academic and commercial labs around the world.

 

LIPA gene analysis is also helpful in the diagnosis of LAL-D, with over 100 LIPA mutations having been identified in patients with LAL deficiency (7). Gene panels for associated diagnoses are becoming available and may allow diagnosis of LAL-D even when clinical awareness is low.

 

DIFFERENTIAL DIAGNOSIS

 

Given the clinical presentation of LAL-D, it is important to consider it in the differential diagnosis of patients presenting with characteristic lipid findings and liver disease. The lipid abnormalities of LAL-D are similar to patients with heterozygous familial hypercholesterolemia (HeFH) and familial combined hypercholesterolemia. A detailed family history may help differentiate the autosomal dominant HeFH from recessive LAL-D. Expert opinion recommends checking liver transaminases in all children and adults before initiating statin therapy (35). LAL-D should be considered in patients with elevated liver enzymes and lipid abnormalities.

 

LAL-D is often mistaken for non-alcoholic fatty liver disease (NAFLD); however, LAL-D is associated with mainly microvesicular steatosis and NAFLD with macrovesicular steatosis.  LAL-D should be included the differential diagnosis of any non-obese patient with hepatic steatosis, as well as patients with unexplained ALT elevations. 

 

MANAGEMENT

 

Disease specific therapy is now available to treat patients with LAL-D. However, prior to the approval of sebelipase alfa (Kanumaâ, Alexion Pharmaceuticals, New Haven, CT), lipid lowering therapy, liver transplant, and stem cell transplant were often tried.

 

HMG-CoA reductase inhibitors have been used to lower LDL-C as well as reduce the risk of atherosclerotic heart disease. The first reported use in a patient with LAL-D was in a 9-year-old girl with elevated LDL-C, low HDL-C, and hepatomegaly with a liver biopsy that showed fibrosis and cirrhosis.  During therapy with lovastatin, lipid parameters improved and the authors showed a reduction in cholesterol synthesis and decreased secretion of apo B-containing lipoproteins (36). However, in a report of three patients treated with lovastatin for 12 months, no significant changes were seen in lipid parameters and liver histology (37). In a review of cases in the literature, 12 patients with LAL-D were treated with HMG CoA reductase inhibitors with multiple liver biopsies. None of the 12 patients had improvement on liver histology, with all 12 patients having progressive liver disease (1). 

 

Both hematopoietic stem cell transplant and liver transplant have been attempted to treat LAL-D, however, neither address the multi-system nature of the disease. Limited information is available about the long-term outcome of patients who have undergone liver transplant (1).

 

Sebelipase alfa, a recombinant human enzyme-replacement, is FDA approved for the treatment of LAL-D (38). The amino acid sequence for sebelipase alfa is the same as that of human LAL.  A multicenter, double-blind, placebo controlled, randomized study in 66 patients analyzed the safety and effectiveness of sebelipase alfa (39). By week 20, patients treated with sebelipase alfa demonstrated a decrease in LDL-C of 28% versus 6% in the placebo group. The treatment group also demonstrated improvement in triglyceride and HDL-C level. Normalization of ALT occurred in 31% of patients in the treatment group versus 7% in the placebo group. This was accompanied by reduction in hepatic fat content assessed by multi-echo gradient echo MRI of 32% in the treatment group versus 4% in the placebo group

 

Table 1. Clinical Trials of Sebelipase Alfa

Study

Subjects

Age

Dose (per kg body weight)

Duration

Reference

LAL-CL01

 

 

9

 

 

 

31.6 ± 10.7 yrs (mean ± SD):

Escalating doses: 0.35, 1, or 3 mg weekly (given to cohort of 3 patients each)

4 wks

(38)

LAL-CL02

66

50 <18 yrs, age range at randomization: 4-58 years

1 mg every other week

Initial 20 wks, followed by an open-label treatment phase for 65 patients

(39)

LAL-CL03

9

3.0 months (median)

Weekly infusions: 0.35 mg x 2 weeks; then 1 mg, with dose increase to 3 mg*

12 months

(40)

LAL-CL04

8

18 to 65 yrs

1 or 3 mg every other week

Through to 52 wks

(41)

*Two infants had dose subsequently increased to 5 mg/kg weekly

Modified from Pastores GM, Hughes DA. Lysosomal Acid Lipase Deficiency: Therapeutic Options. Drug Des Devel Ther. 2020 Feb 11;14:591-601.

 

The frequency and distribution of adverse events were similar in the treatment and placebo group, and most adverse events were considered unrelated to the study drug (Table 2) (39). Clinical trials have shown that 3/106 patients experienced reactions consistent with anaphylaxis during infusion, occurring as early as the sixth infusion and as late as 1 year. Twenty percent (21/106) of patients experienced symptoms consistent with hypersensitivity reaction during or within 4 hours of completion of the infusion (38-41). The current dosing recommendation from the manufacturer for infantile-onset LAL-D is 1mg/kg IV weekly with escalation to 3mg/kg weekly in those who do not achieve appropriate clinical response.  For child and adults presenting with LAL-D, the recommended dose is 1 mg/kg every other week (38). Further long-term follow-up studies are needed. 

 

Table 2. Adverse Events with Sebelipase Alfa

Event

Sebelipase Alfa (N=36)

Placebo (N=30)

Any adverse event

31 (86%)

28 (93%)

Gastrointestinal events1

18 (50%)

12 (40%)

Headache

10 (28%)

6 (20%)

Fever

7 (19%)

6 (20%)

Oropharyngeal pain

6 (17%)

1 (3%)

Upper respiratory tract infection

6 (17%)

6 (20%)

Epistaxis

4 (11%)

3 (10%)

Asthenia

3 (8%)

1 (3%)

Cough

3 (8%)

3 (10%)

Adapted from Burton, et al., NEJM 2015

1Gastrointestinal adverse events (diarrhea, abdominal pain, constipation, nausea, vomiting)

 

In contrast to survival rates of <12 months in infants with rapidly progressive LAL-D, results of two open-label studies of enzyme replacement therapy with sebelipase alfa, VITAL (NCT01371825) and CL08 (NCT02193867), in 19 infants reported prolonged survival to 12 months (79%) and 5 years of age (68%) in the combined population. The median age of surviving patients was 5.2 (VITAL) and 3.2 years (CL08). In both studies, median weight-for-age, length-for-age, and mid-upper arm circumference-for-age Z-scores increased from baseline to end of study, and decreases in median liver and spleen volume were observed. No patient discontinued treatment because of treatment-emergent adverse events. Infusion-associated reactions (94% in VITAL and 88% in CL08) were mild or moderate in severity (42).

 

In older children (>4 years) and adults with LAL-D, a phase III randomized study of sebelipase alfa (RISE, NCT01757184) included a 20-week, double-blind, placebo-controlled period; a 130-week, open-label, extension period; and a 104-week, open-label, expanded treatment period. 59/66 patients completed the study. The study found that early and rapid improvements in markers of liver injury and lipid abnormalities with sebelipase alfa were sustained, with no progression of liver disease, for up to 5 years (43).

 

CONCLUSION

 

Consensus recommendations for the initial assessment and ongoing monitoring of children and adults with LAL deficiency have been published to help improve the management of infants, children and adults with confirmed LAL-D (Figures 4 and 5) (44).

 

Figure 4. Recommendations for Baseline Assessment of Children and Adults with LAL Deficiency.

Figure 5. Schedule of Ongoing Monitoring of Adults and Children with LAL Deficiency.

 

REFERENCES

 

  1. Bernstein DL, Hulkova H, Bialer MG, Desnick RJ. Cholesteryl ester storage disease: review of the findings in 135 reported patients with an underdiagnosed disease. J Hepatol 2013; 58:1230-1243
  2. Gregory A. Grabowski LC, H Du. Acid lipase deficiency: Wolman disease and cholesteryl ester storage disease. New York: McGraw-Hill, Inc.
  3. Abramov A, Schorr S, Wolman M. Generalized xanthomatosis with calcified adrenals. AMA J Dis Child 1956; 91:282-286
  4. Fredrickson DS, Sloan HR, Ferrans VJ, Demosky SJ, Jr. Cholesteryl ester storage disease: a most unusual manifestation of deficiency of two lysosomal enzyme activities. Trans Assoc Am Physicians 1972; 85:109-119
  5. Infante R, Polonovski J, Caroli J. [Cholesterolic polycoria in adults. II. Biochemical study]. Presse Med (1893)1967; 75:2829-2832
  6. Schiff L, Schubert WK, McAdams AJ, Spiegel EL, O'Donnell JF. Hepatic cholesterol ester storage disease, a familial disorder. I. Clinical aspects. Am J Med 1968; 44:538-546
  7. Pisciotta L, Tozzi G, Travaglini L, Taurisano R, Lucchi T, Indolfi G, Papadia F, Di Rocco M, D'Antiga L, Crock P, Vora K, Nightingale S, Michelakakis H, Garoufi A, Lykopoulou L, Bertolini S, Calandra S. Molecular and clinical characterization of a series of patients with childhood-onset lysosomal acid lipase deficiency. Retrospective investigations, follow-up and detection of two novel LIPA pathogenic variants. Atherosclerosis 2017; 265:124-132
  8. Kyriakides EC, Filippone N, Paul B, Grattan W, Balint JA. LIPID STUDIES IN WOLMAN'S DISEASE. Pediatrics 1970; 46:431-436
  9. Yoshida H, Kuriyama M. Genetic lipid storage disease with lysosomal acid lipase deficiency in rats. Lab Anim Sci 1990; 40:486-489
  10. Goldstein JL, Dana SE, Faust JR, Beaudet AL, Brown MS. Role of lysosomal acid lipase in the metabolism of plasma low density lipoprotein. Observations in cultured fibroblasts from a patient with cholesteryl ester storage disease. Journal of Biological Chemistry 1975; 250:8487-8495
  11. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Journal of Clinical Investigation 2002; 109:1125-1131
  12. Jeon T-I, Osborne TF. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol Metab2012; 23:65-72
  13. Brown MS, Sobhani MK, Brunschede GY, Goldstein JL. Restoration of a regulatory response to low density lipoprotein in acid lipase-deficient human fibroblasts. Journal of Biological Chemistry 1976; 251:3277-3286
  14. Cummings MH, Watts GF. Increased hepatic secretion of very-low-density lipoprotein apolipoprotein B-100 in cholesteryl ester storage disease. Clinical Chemistry 1995; 41:111-114
  15. Sando GN, Ma GP, Lindsley KA, Wei YP. Intercellular transport of lysosomal acid lipase mediates lipoprotein cholesteryl ester metabolism in a human vascular endothelial cell-fibroblast coculture system. Cell Regul 1990; 1:661-674
  16. Bowden KL, Bilbey NJ, Bilawchuk LM, Boadu E, Sidhu R, Ory DS, Du H, Chan T, Francis GA. Lysosomal acid lipase deficiency impairs regulation of ABCA1 gene and formation of high density lipoproteins in cholesteryl ester storage disease. J Biol Chem 2011; 286:30624-30635
  17. Muntoni S, Wiebusch H, Jansen-Rust M, Rust S, Seedorf U, Schulte H, Berger K, Funke H, Assmann G. Prevalence of Cholesteryl Ester Storage Disease. Arteriosclerosis, Thrombosis, and Vascular Biology 2007; 27:1866-1868
  18. Lohse P, Maas S, Lohse P, Elleder M, Kirk JM, Besley GTN, Seidel D. Compound heterozygosity for a Wolman mutation is frequent among patients with cholesteryl ester storage disease. Journal of Lipid Research 2000; 41:23-31
  19. Reiner Ž, Guardamagna O, Nair D, Soran H, Hovingh K, Bertolini S, Jones S, Ćorić M, Calandra S, Hamilton J, Eagleton T, Ros E. Lysosomal acid lipase deficiency – An under-recognized cause of dyslipidaemia and liver dysfunction. Atherosclerosis 2014; 235:21-30
  20. Valles-Ayoub Y, Esfandiarifard S, No D, Sinai P, Khokher Z, Kohan M, Kahen T, Darvish D. Wolman Disease (LIPA p.G87V) Genotype Frequency in People of Iranian-Jewish Ancestry. Genetic Testing and Molecular Biomarkers 2011; 15:395-398
  21. Reynolds TM, Mewies C, Hamilton J, Wierzbicki AS, group PPC. Identification of rare diseases by screening a population selected on the basis of routine pathology results-the PATHFINDER project: lysosomal acid lipase/cholesteryl ester storage disease substudy. J Clin Pathol 2018; 71:608-613
  22. Wolman M, Sterk VV, Gatt S, Frenkel M. PRIMARY FAMILIAL XANTHOMATOSIS WITH INVOLVEMENT AND CALCIFICATION OF THE ADRENALS. Pediatrics 1961; 28:742-757
  23. Jones SA, Valayannopoulos V, Schneider E, Eckert S, Banikazemi M, Bialer M, Cederbaum S, Chan A, Dhawan A, Di Rocco M, Domm J, Enns GM, Finegold D, Gargus JJ, Guardamagna O, Hendriksz C, Mahmoud IG, Raiman J, Selim LA, Whitley CB, Zaki O, Quinn AG. Rapid progression and mortality of lysosomal acid lipase deficiency presenting in infants. Genet Med 2016; 18:452-458
  24. Zhang B, Porto AF. Cholesteryl Ester Storage Disease. Journal of Pediatric Gastroenterology &amp; Nutrition2013; 56:682-685
  25. Tebani A, Sudrie-Arnaud B, Boudabous H, Brassier A, Anty R, Snanoudj S, Abergel A, Abi Warde MT, Bardou-Jacquet E, Belbouab R, Blanchet E, Borderon C, Bronowicki JP, Cariou B, Carette C, Dabbas M, Dranguet H, de Ledinghen V, Ferrieres J, Guillaume M, Krempf M, Lacaille F, Larrey D, Leroy V, Musikas M, Nguyen-Khac E, Ouzan D, Perarnau JM, Pilon C, Ratzlu V, Thebaut A, Thevenot T, Tragin I, Triolo V, Verges B, Vergnaud S, Bekri S. Large-scale screening of lipase acid deficiency in at risk population. Clin Chim Acta 2021; 519:64-69
  26. Pericleous M, Kelly C, Wang T, Livingstone C, Ala A. Wolman's disease and cholesteryl ester storage disorder: the phenotypic spectrum of lysosomal acid lipase deficiency. Lancet Gastroenterol Hepatol 2017; 2:670-679
  27. Maciejko JJ. Managing Cardiovascular Risk in Lysosomal Acid Lipase Deficiency. Am J Cardiovasc Drugs2017; 17:217-231
  28. Elleder M, Chlumska A, Hyanek J, Poupetova H, Ledvinova J, Maas S, Lohse P. Subclinical course of cholesteryl ester storage disease in an adult with hypercholesterolemia, accelerated atherosclerosis, and liver cancer. J Hepatol 2000; 32:528-534
  29. Wilson DP, Friedman M, Marulkar S, Hamby T, Bruckert E. Sebelipase alfa improves atherogenic biomarkers in adults and children with lysosomal acid lipase deficiency. J Clin Lipidol 2018; 12:604-614
  30. Burton BK, Deegan PB, Enns GM, Guardamagna O, Horslen S, Hovingh GK, Lobritto SJ, Malinova V, McLin VA, Raiman J, Di Rocco M, Santra S, Sharma R, Sykut-Cegielska J, Whitley CB, Eckert S, Valayannopoulos V, Quinn AG. Clinical Features of Lysosomal Acid Lipase Deficiency. J Pediatr Gastroenterol Nutr 2015; 61:619-625
  31. Kostner GM, Hadorn B, Roscher A, Zechner R. Plasma lipids and lipoproteins of a patient with cholesteryl ester storage disease. Journal of Inherited Metabolic Disease 1984; 8:9-12
  32. Ambler GK, Hoare M, Brais R, Shaw A, Butler A, Flynn P, Deegan P, Griffiths WJH. Orthotopic liver transplantation in an adult with cholesterol ester storage disease. JIMD Rep 2013; 8:41-46
  33. Hůlková H, Elleder M. Distinctive histopathological features that support a diagnosis of cholesterol ester storage disease in liver biopsy specimens. Histopathology 2012; 60:1107-1113
  34. Hamilton J, Jones I, Srivastava R, Galloway P. A new method for the measurement of lysosomal acid lipase in dried blood spots using the inhibitor Lalistat 2. Clinica Chimica Acta 2012; 413:1207-1210
  35. Bays H, Cohen DE, Chalasani N, Harrison Stephen A. An assessment by the Statin Liver Safety Task Force: 2014 update. Journal of Clinical Lipidology 2014; 8:S47-S57
  36. Ginsberg HN, Le NA, Short MP, Ramakrishnan R, Desnick RJ. Suppression of apolipoprotein B production during treatment of cholesteryl ester storage disease with lovastatin. Implications for regulation of apolipoprotein B synthesis. J Clin Invest 1987; 80:1692-1697
  37. Di Bisceglie AM, Ishak KG, Rabin L, Hoeg JM. Cholesteryl ester storage disease: Hepatopathology and effects of therapy with lovastatin. Hepatology 1990; 11:764-772
  38. Balwani M, Breen C, Enns GM, Deegan PB, Honzík T, Jones S, Kane JP, Malinova V, Sharma R, Stock EO, Valayannopoulos V, Wraith JE, Burg J, Eckert S, Schneider E, Quinn AG. Clinical effect and safety profile of recombinant human lysosomal acid lipase in patients with cholesteryl ester storage disease. Hepatology 2013; 58:950-957
  39. Burton BK, Balwani M, Feillet F, Barić I, Burrow TA, Camarena Grande C, Coker M, Consuelo-Sánchez A, Deegan P, Di Rocco M, Enns GM, Erbe R, Ezgu F, Ficicioglu C, Furuya KN, Kane J, Laukaitis C, Mengel E, Neilan EG, Nightingale S, Peters H, Scarpa M, Schwab KO, Smolka V, Valayannopoulos V, Wood M, Goodman Z, Yang Y, Eckert S, Rojas-Caro S, Quinn AG. A Phase 3 Trial of Sebelipase Alfa in Lysosomal Acid Lipase Deficiency. New England Journal of Medicine 2015; 373:1010-1020
  40. Jones SA, Rojas-Caro S, Quinn AG, Friedman M, Marulkar S, Ezgu F, Zaki O, Gargus JJ, Hughes J, Plantaz D, Vara R, Eckert S, Arnoux JB, Brassier A, Le Quan Sang KH, Valayannopoulos V. Survival in infants treated with sebelipase Alfa for lysosomal acid lipase deficiency: an open-label, multicenter, dose-escalation study. Orphanet J Rare Dis 2017; 12:25
  41. Valayannopoulos V, Malinova V, Honzík T, Balwani M, Breen C, Deegan PB, Enns GM, Jones SA, Kane JP, Stock EO, Tripuraneni R, Eckert S, Schneider E, Hamilton G, Middleton MS, Sirlin C, Kessler B, Bourdon C, Boyadjiev SA, Sharma R, Twelves C, Whitley CB, Quinn AG. Sebelipase alfa over 52 weeks reduces serum transaminases, liver volume and improves serum lipids in patients with lysosomal acid lipase deficiency. Journal of hepatology 2014; 61:1135-1142
  42. Vijay S, Brassier A, Ghosh A, Fecarotta S, Abel F, Marulkar S, Jones SA. Long-term survival with sebelipase alfa enzyme replacement therapy in infants with rapidly progressive lysosomal acid lipase deficiency: final results from 2 open-label studies. Orphanet J Rare Dis 2021; 16:13
  43. Burton BK, Feillet F, Furuya KN, Marulkar S, Balwani M. Sebelipase alfa in children and adults with lysosomal acid lipase deficiency: Final results of the ARISE study. J Hepatol 2022; 76:577-587
  44. Kohli R, Ratziu V, Fiel MI, Waldmann E, Wilson DP, Balwani M. Initial assessment and ongoing monitoring of lysosomal acid lipase deficiency in children and adults: Consensus recommendations from an international collaborative working group. Molecular Genetics and Metabolism 2020; 129:59-66

Lysosomal Acid Lipase Deficiency

ABSTRACT

 

Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive genetic disease with variable presentation which often leads to severe morbidity and mortality. More than 100 LIPA loss of function mutations have been identified, the most common reported mutation being a splice junction mutation in exon 8. The true prevalence of the disease is unknown, but is estimated to be between 1:40,000 to 1:300,000. Infantile-onset LAL-D is generally fatal within the first 12 months of life. Common presenting symptoms in the late-onset form include dyslipidemia (elevated LDL-C, low HDL-C), elevated liver transaminases, hepatomegaly, and splenomegaly.  Prior to the availability of enzyme-replacement therapy, individuals with LAL-D were treated with lipid lowering medication, liver transplant, and stem cell transplant, none of which corrected the multisystem nature of the disorder. Sebelipase alfa (Kanuma®), a recombinant human lysosomal acid lipase, was approved by the FDA in 2015 to treat LAL-D. Phase 3 studies have shown an improvement in lipid parameters and liver enzymes. Long term studies demonstrating the safety and efficacy of sebelipase alfa in infants, children and adults are ongoing.

 

INTRODUCTION

 

Lysosomal acid lipase deficiency (LAL-D) is a rare, heterogeneous, autosomal recessive genetic disease, the manifestations of which include a clinical continuum. LAL-D is characterized by accumulation of cholesteryl esters and triglycerides primarily in the liver and spleen, but with involvement of other organs as well. Clinically, LAL-D is under-recognized, leading to a delay in diagnosis. It is often mistaken for more common conditions with similar clinical and laboratory findings, such as heterozygous familial hypercholesterolemia (FH) and non-alcoholic fatty liver disease (NAFLD) (1,2). Correct diagnosis and timely intervention are critical to prolonging life and improving outcomes.

 

Similar to other lysosomal storage disorders, LAL-D presents across a clinical spectrum from infancy to adulthood. Historically, affected infants who presented within the first year of life were known as Wolman Disease while those who symptoms were delayed until childhood were referred to as cholesteryl ester storage disease [CESD]. Wolman disease, which has a rapidly progressive course, was first described in 1956. Affected infants have severe malnutrition, adrenal calcifications, hepatosplenomegaly, and death within the first few months of life (3). In contrast, CESD is seen as having a variable clinical spectrum with recognition of the disorder occurring from childhood into adulthood. Fredrickson, Schiff, Langeron, and Infante were the first to describe CESD in individuals with presentation from the first to fourth decades of life, and noted them to be less severe than those described by Wolman (4-6).

 

INHERITANCE AND GENETICS

 

LAL-D is an autosomal recessive disease that arises from mutations at the LAL locus on chromosome 10q23.2.  Affected individuals are either homozygous or compound heterozygous for LIPA mutations, with more than 100 LIPA mutations having been identified (7).

 

Lysosomal acid lipase (LAL) plays a central role in intracellular lipid metabolism (8,9). LAL is the only lipase contained within lysosomes that hydrolyzes cholesteryl esters and triglycerides.  After cleavage by LAL, free cholesterol and fatty acids exit the lysosome to enter the cytosol (Figure 1). These cleaved products play an important role in cholesterol homeostasis. Free cholesterol interacts with transcription factors (sterol regulatory element binding proteins [SREBPs]) to modulate production of intracellular cholesterol. As intracellular free cholesterol increases, there is a down regulation of LDL receptors mediated by SREBP-2, resulting in less LDL entering the cell. Additionally, there is inhibition of hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase, resulting in decreased cholesterol production, as well as stimulation of acyl-cholesterol acyltransferase (leading to increased cholesterol esterification). Finally, increased intracellular fatty acid leads to inhibition of triglyceride and phospholipid production and decreased fatty acid synthesis (10-12).

 

Deficiency of LAL results in diminished or absent hydrolysis of cholesteryl esters and triglycerides, trapping cholesterol esters and TG within the lysosome. This results in a decrease in cytosolic free cholesterol and a compensatory, upregulation in the cholesterol synthetic pathway (HMG CoA reductase activity) and endocytosis via increased LDL receptors. There is increased production of apolipoprotein B and very low-density lipoprotein (VLDL-C) (13-15). The dysregulated expression of the LDL-cholesterol-dependent ATP binding cassette transporter 1 (ABCA1), similar to that seen in Niemann-Pick type C1, results in decreased levels of HDL-C (16). The characteristic dyslipidemia seen in individuals with LAL-D includes elevated total cholesterol, elevated LDL-C, and low HDL-C (2).

 

Figure 1. Cellular Cholesterol Homeostasis in Heathy Individuals and Patients with LAL-D

The true incidence of LAL-D is unknown. Estimates suggest overall disease prevalence between 1:40,000 to 1:300,000, depending on ethnicity and geographical location (1,2,17). The most commonly inherited defect is a splice junction mutation in exon 8, E8SJM (c.894G>A). It is assumed that 50-70% of adults and children with LAL-D have E8SJM (17,18). Studies in the general population have shown that the estimated frequency of E8SJM allele is 0.0013 in Caucasians, 0.0017 in US Hispanics, 0.0010 in US Ashkenazi Jews, and 0.0005 in Asians (19).  Population screening for E8SJM among healthy West German individuals reveal a heterozygote frequency of ~ 1:200 individuals. Jewish infants of Iraqi or Iranian origin appear to be at high risk for LAL-D with an estimated incidence of 1:4,200 in the Los Angeles community (20).

 

A study attempting to identify the prevalence of LAL-D from patients with abnormal results in laboratory databases (elevated LDL-C and abnormalities on liver tests) identified a total of 1825 patients who subsequently underwent a dried blood spot sample for determination of LAL enzyme activity. No cases of LAL-D were identified. The results of this study demonstrate the potential of databases in helping to identify patients with specific patterns of results to allow targeted testing for possible causes of disease. Biochemical screening suggested that the gene frequency of LAL deficiency in adults is less than 1:100 (21). Additionally, histopathology databases of liver biopsies were analyzed searching for patients with features of 'microvesicular cirrhosis' or 'cryptogenic cirrhosis'. DNA was available from six patients and two were homozygous for LAL c.46A>C;p.Thr16Pro, an unclassified variant in exon 2 (21). The results of these studies suggest the potential of databases in helping to identify patients with specific laboratory results or those who had certain biopsy findings to allow targeted testing for possible causes of disease.

 

PRESENTATION

 

The symptoms of LAL-D are quite varied, and are related to the age that clinical manifestations first appear (Figure 2).  Individuals who present within the first few days to first month of life often have vomiting, diarrhea, hepatosplenomegaly, abdominal distention, and severe failure to thrive. The first symptom observed is usually vomiting, which has been described as forceful and persistent. Accompanying these symptoms are usually watery diarrhea and low-grade fever. Symptoms generally persist despite multiple medical interventions and may lead to severe malnutrition. A hallmark of infantile-onset LAL-D is adrenal enlargement and calcification, often seen on imaging, but not required for diagnosis. Calcifications of the adrenal gland as well as adrenal insufficiency have been documented. Few patients survive beyond 12 months of age (2,3,22), with those that have growth failure often dying by four months of age (23).

 

In contrast, the clinical presentation and progression of LAL-D can be variable in older children and adults. However, there are common clinical manifestations that have been reported in this group of patients. In a review of 71 patients two thirds presented with their first symptoms before the age of 5 years. Hepatomegaly was present in all the patients; 86% had splenomegaly.  Gastrointestinal symptoms were present in 30% and included vomiting and diarrhea [18%], failure to thrive [16%], abdominal pain [10%], gastrointestinal bleeding [8%], and gallbladder disease [4%]. Elevation of cholesterol was present in 90% (24). In a separate review of 135 patients, the median age of onset of symptoms was 5 years with a range from birth to 68 years.  Hepatomegaly was present in 99.3% of patients. The most common extrahepatic findings were steatorrhea, poor growth, gallbladder dysfunction, and cardiovascular disease. Total cholesterol was elevated in all 110 patients (1). 

 

The disease severity is likely dependent on the efficiency of alternative pathways, but not on the level of residual enzyme activity (25). In adults, the most frequent symptoms are abdominal pain, hepatomegaly, and laboratory abnormalities that include increased levels of transaminases and cholesterol. Differential diagnostic considerations include autoimmune hepatitis, NASH, alpha1-antitrypsin deficiency, and Wilson disease. Of concern is the potential for premature atherosclerosis in affected individuals.  Although the occurrence of cardiovascular events has not been extensively studied, case reports and observational studies have documented the presence of arterial plaque and atheroma at a very early age (26-28). As a result, many patients with this disease have been prescribed lipid-lowering medications (1). While lipid lowering in the setting of LAL-A has been variable, statins increase hepatic uptake of LDL and, as a consequence, may worsen the lipid overload (29). It is important to note that seven asymptomatic adults, diagnosed in the third to sixth decade of life, have been reported.  All were coincidently found to have confirmed LAL-D, yet none had detectable hepatomegaly (28). 

 

The most consistent biochemical abnormalities seen in late onset LAL-D include elevated liver transaminases and plasma lipids. In a study of 49 patients designed to characterize clinical manifestations of LAL-D, mean ALT, AST, and GGT were 92.4, 87.8, and 52.2 U/L at the first measurement. In this study elevated GGT levels were uncommon (only 20% had values > 40 U/L) (30). In another study, liver dysfunction occurred in 100% of 135 patients and 73% of the 11 reported deaths were due to liver failure (1). Mean LDL-C at the time of first measurement was 202.9 mg/dL, and reported as abnormal in 64.4% of patients. Mean total cholesterol was 269.5 mg/dL and was abnormal in 62.5%. Mean HDL-C was 37.5 mg/dL and abnormal in 43.5% of patients (30). The lipid abnormalities seen most closely resemble type II-b dyslipidemia (31).  Although elevated LDL-C seems to be a feature of LAL-D, it remains unclear whether or not LAL-D is a cause of early atherosclerosis. Case reports and several autopsy studies have noted aortic stenosis and found narrowing of the coronary artery secondary to atheromatous plaque in patients with LAL-D (2,32).

 

Figure 2. Clinical Presentation of LAL-D

 

On gross examination, the liver of patients with LAL-D is enlarged and appears greasy. Liver biopsies in paraffin sections have a predominance of microvesicular steatosis, which is uniform.  Microvesicular steatosis, per se, is not pathognomonic of LAL-D, being found in other liver diseases as well. Foamy macrophages, containing lipid and ceroid, are present in the sinusoids and portal tracts (Figure 3).  Staining for LAMP1, LAMP2, and LIMP2, or with a lysosomal luminal protein (cathepsin D), can assist identifying lipid accumulation as lysosomal, may help differentiate LAL-D from other causes of microvesicular steatosis. Another pathognomonic feature of LAL-D is birefringent cholesterol ester crystals in hepatocytes and Kupffer cells, using polarized light on electron microscopy. The liver disease generally progresses to fibrosis followed by micronodular cirrhosis (1,2,33).

 

Figure 3. Liver Biopsies in Patients with LAL-D. A) Image of the portal tract and hepatocytes with mainly microvesicular steatosis. With microvesicular steatosis, the fat does not cause the nucleus to be pushed out to the side. B) Larger magnification of the portal tract. FM points to the foamy appearing cytoplasm, these are macrophages with something being stored in them. GC is pointing to a giant cell.

 

DIAGNOSTIC TESTS

 

LAL-D can be diagnosed by demonstrating deficient LAL enzyme activity, as well as by genetic testing identifying mutations of the LIPA gene. Historically, enzyme activity was measured in cultured fibroblasts, peripheral leukocytes, or liver tissue. Various lipase substrates, which were not specific for LAL, were used. In the review by Bernstein, enzyme activities were reported in 114 patients and ranged from undetectable to 16% of normal, with values for most patients being between <1%-10%. However, given assay variability, residual enzyme activity is not predictive of disease severity nor can it be compared from one lab to another (1).  

 

A newer method has been developed to determine LAL activity. This method measures LAL activity in dried blood spots (DBS), and uses Lalistat 2, a highly specific inhibitor of LAL. LAL activity is determined by comparing total lipase activity to lipase activity with Lalistat 2. This method is able to differentiate normal from affected individuals. This DBS technique has advantages over the fibroblast/peripheral leukocyte based test including small sample size, the ability to transport the specimen to the testing facility at ambient temperature, and sample stability (34). This blood test is available at a number of academic and commercial labs around the world.

 

LIPA gene analysis is also helpful in the diagnosis of LAL-D, with over 100 LIPA mutations having been identified in patients with LAL deficiency (7). Gene panels for associated diagnoses are becoming available and may allow diagnosis of LAL-D even when clinical awareness is low.

 

DIFFERENTIAL DIAGNOSIS

 

Given the clinical presentation of LAL-D, it is important to consider it in the differential diagnosis of patients presenting with characteristic lipid findings and liver disease. The lipid abnormalities of LAL-D are similar to patients with heterozygous familial hypercholesterolemia (HeFH) and familial combined hypercholesterolemia. A detailed family history may help differentiate the autosomal dominant HeFH from recessive LAL-D. Expert opinion recommends checking liver transaminases in all children and adults before initiating statin therapy (35). LAL-D should be considered in patients with elevated liver enzymes and lipid abnormalities.

 

LAL-D is often mistaken for non-alcoholic fatty liver disease (NAFLD); however, LAL-D is associated with mainly microvesicular steatosis and NAFLD with macrovesicular steatosis.  LAL-D should be included the differential diagnosis of any non-obese patient with hepatic steatosis, as well as patients with unexplained ALT elevations. 

 

MANAGEMENT

 

Disease specific therapy is now available to treat patients with LAL-D. However, prior to the approval of sebelipase alfa (Kanumaâ, Alexion Pharmaceuticals, New Haven, CT), lipid lowering therapy, liver transplant, and stem cell transplant were often tried.

 

HMG-CoA reductase inhibitors have been used to lower LDL-C as well as reduce the risk of atherosclerotic heart disease. The first reported use in a patient with LAL-D was in a 9-year-old girl with elevated LDL-C, low HDL-C, and hepatomegaly with a liver biopsy that showed fibrosis and cirrhosis.  During therapy with lovastatin, lipid parameters improved and the authors showed a reduction in cholesterol synthesis and decreased secretion of apo B-containing lipoproteins (36). However, in a report of three patients treated with lovastatin for 12 months, no significant changes were seen in lipid parameters and liver histology (37). In a review of cases in the literature, 12 patients with LAL-D were treated with HMG CoA reductase inhibitors with multiple liver biopsies. None of the 12 patients had improvement on liver histology, with all 12 patients having progressive liver disease (1). 

 

Both hematopoietic stem cell transplant and liver transplant have been attempted to treat LAL-D, however, neither address the multi-system nature of the disease. Limited information is available about the long-term outcome of patients who have undergone liver transplant (1).

 

Sebelipase alfa, a recombinant human enzyme-replacement, is FDA approved for the treatment of LAL-D (38). The amino acid sequence for sebelipase alfa is the same as that of human LAL.  A multicenter, double-blind, placebo controlled, randomized study in 66 patients analyzed the safety and effectiveness of sebelipase alfa (39). By week 20, patients treated with sebelipase alfa demonstrated a decrease in LDL-C of 28% versus 6% in the placebo group. The treatment group also demonstrated improvement in triglyceride and HDL-C level. Normalization of ALT occurred in 31% of patients in the treatment group versus 7% in the placebo group. This was accompanied by reduction in hepatic fat content assessed by multi-echo gradient echo MRI of 32% in the treatment group versus 4% in the placebo group

 

Table 1. Clinical Trials of Sebelipase Alfa

Study

Subjects

Age

Dose (per kg body weight)

Duration

Reference

LAL-CL01

 

 

9

 

 

 

31.6 ± 10.7 yrs (mean ± SD):

Escalating doses: 0.35, 1, or 3 mg weekly (given to cohort of 3 patients each)

4 wks

(38)

LAL-CL02

66

50 <18 yrs, age range at randomization: 4-58 years

1 mg every other week

Initial 20 wks, followed by an open-label treatment phase for 65 patients

(39)

LAL-CL03

9

3.0 months (median)

Weekly infusions: 0.35 mg x 2 weeks; then 1 mg, with dose increase to 3 mg*

12 months

(40)

LAL-CL04

8

18 to 65 yrs

1 or 3 mg every other week

Through to 52 wks

(41)

*Two infants had dose subsequently increased to 5 mg/kg weekly

Modified from Pastores GM, Hughes DA. Lysosomal Acid Lipase Deficiency: Therapeutic Options. Drug Des Devel Ther. 2020 Feb 11;14:591-601.

 

The frequency and distribution of adverse events were similar in the treatment and placebo group, and most adverse events were considered unrelated to the study drug (Table 2) (39). Clinical trials have shown that 3/106 patients experienced reactions consistent with anaphylaxis during infusion, occurring as early as the sixth infusion and as late as 1 year. Twenty percent (21/106) of patients experienced symptoms consistent with hypersensitivity reaction during or within 4 hours of completion of the infusion (38-41). The current dosing recommendation from the manufacturer for infantile-onset LAL-D is 1mg/kg IV weekly with escalation to 3mg/kg weekly in those who do not achieve appropriate clinical response.  For child and adults presenting with LAL-D, the recommended dose is 1 mg/kg every other week (38). Further long-term follow-up studies are needed. 

 

Table 2. Adverse Events with Sebelipase Alfa

Event

Sebelipase Alfa (N=36)

Placebo (N=30)

Any adverse event

31 (86%)

28 (93%)

Gastrointestinal events1

18 (50%)

12 (40%)

Headache

10 (28%)

6 (20%)

Fever

7 (19%)

6 (20%)

Oropharyngeal pain

6 (17%)

1 (3%)

Upper respiratory tract infection

6 (17%)

6 (20%)

Epistaxis

4 (11%)

3 (10%)

Asthenia

3 (8%)

1 (3%)

Cough

3 (8%)

3 (10%)

Adapted from Burton, et al., NEJM 2015

1Gastrointestinal adverse events (diarrhea, abdominal pain, constipation, nausea, vomiting)

 

In contrast to survival rates of <12 months in infants with rapidly progressive LAL-D, results of two open-label studies of enzyme replacement therapy with sebelipase alfa, VITAL (NCT01371825) and CL08 (NCT02193867), in 19 infants reported prolonged survival to 12 months (79%) and 5 years of age (68%) in the combined population. The median age of surviving patients was 5.2 (VITAL) and 3.2 years (CL08). In both studies, median weight-for-age, length-for-age, and mid-upper arm circumference-for-age Z-scores increased from baseline to end of study, and decreases in median liver and spleen volume were observed. No patient discontinued treatment because of treatment-emergent adverse events. Infusion-associated reactions (94% in VITAL and 88% in CL08) were mild or moderate in severity (42).

 

In older children (>4 years) and adults with LAL-D, a phase III randomized study of sebelipase alfa (RISE, NCT01757184) included a 20-week, double-blind, placebo-controlled period; a 130-week, open-label, extension period; and a 104-week, open-label, expanded treatment period. 59/66 patients completed the study. The study found that early and rapid improvements in markers of liver injury and lipid abnormalities with sebelipase alfa were sustained, with no progression of liver disease, for up to 5 years (43).

 

CONCLUSION

 

Consensus recommendations for the initial assessment and ongoing monitoring of children and adults with LAL deficiency have been published to help improve the management of infants, children and adults with confirmed LAL-D (Figures 4 and 5) (44).

 

Figure 4. Recommendations for Baseline Assessment of Children and Adults with LAL Deficiency.

Figure 5. Schedule of Ongoing Monitoring of Adults and Children with LAL Deficiency.

 

REFERENCES

 

  1. Bernstein DL, Hulkova H, Bialer MG, Desnick RJ. Cholesteryl ester storage disease: review of the findings in 135 reported patients with an underdiagnosed disease. J Hepatol 2013; 58:1230-1243
  2. Gregory A. Grabowski LC, H Du. Acid lipase deficiency: Wolman disease and cholesteryl ester storage disease. New York: McGraw-Hill, Inc.
  3. Abramov A, Schorr S, Wolman M. Generalized xanthomatosis with calcified adrenals. AMA J Dis Child 1956; 91:282-286
  4. Fredrickson DS, Sloan HR, Ferrans VJ, Demosky SJ, Jr. Cholesteryl ester storage disease: a most unusual manifestation of deficiency of two lysosomal enzyme activities. Trans Assoc Am Physicians 1972; 85:109-119
  5. Infante R, Polonovski J, Caroli J. [Cholesterolic polycoria in adults. II. Biochemical study]. Presse Med (1893)1967; 75:2829-2832
  6. Schiff L, Schubert WK, McAdams AJ, Spiegel EL, O'Donnell JF. Hepatic cholesterol ester storage disease, a familial disorder. I. Clinical aspects. Am J Med 1968; 44:538-546
  7. Pisciotta L, Tozzi G, Travaglini L, Taurisano R, Lucchi T, Indolfi G, Papadia F, Di Rocco M, D'Antiga L, Crock P, Vora K, Nightingale S, Michelakakis H, Garoufi A, Lykopoulou L, Bertolini S, Calandra S. Molecular and clinical characterization of a series of patients with childhood-onset lysosomal acid lipase deficiency. Retrospective investigations, follow-up and detection of two novel LIPA pathogenic variants. Atherosclerosis 2017; 265:124-132
  8. Kyriakides EC, Filippone N, Paul B, Grattan W, Balint JA. LIPID STUDIES IN WOLMAN'S DISEASE. Pediatrics 1970; 46:431-436
  9. Yoshida H, Kuriyama M. Genetic lipid storage disease with lysosomal acid lipase deficiency in rats. Lab Anim Sci 1990; 40:486-489
  10. Goldstein JL, Dana SE, Faust JR, Beaudet AL, Brown MS. Role of lysosomal acid lipase in the metabolism of plasma low density lipoprotein. Observations in cultured fibroblasts from a patient with cholesteryl ester storage disease. Journal of Biological Chemistry 1975; 250:8487-8495
  11. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Journal of Clinical Investigation 2002; 109:1125-1131
  12. Jeon T-I, Osborne TF. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol Metab2012; 23:65-72
  13. Brown MS, Sobhani MK, Brunschede GY, Goldstein JL. Restoration of a regulatory response to low density lipoprotein in acid lipase-deficient human fibroblasts. Journal of Biological Chemistry 1976; 251:3277-3286
  14. Cummings MH, Watts GF. Increased hepatic secretion of very-low-density lipoprotein apolipoprotein B-100 in cholesteryl ester storage disease. Clinical Chemistry 1995; 41:111-114
  15. Sando GN, Ma GP, Lindsley KA, Wei YP. Intercellular transport of lysosomal acid lipase mediates lipoprotein cholesteryl ester metabolism in a human vascular endothelial cell-fibroblast coculture system. Cell Regul 1990; 1:661-674
  16. Bowden KL, Bilbey NJ, Bilawchuk LM, Boadu E, Sidhu R, Ory DS, Du H, Chan T, Francis GA. Lysosomal acid lipase deficiency impairs regulation of ABCA1 gene and formation of high density lipoproteins in cholesteryl ester storage disease. J Biol Chem 2011; 286:30624-30635
  17. Muntoni S, Wiebusch H, Jansen-Rust M, Rust S, Seedorf U, Schulte H, Berger K, Funke H, Assmann G. Prevalence of Cholesteryl Ester Storage Disease. Arteriosclerosis, Thrombosis, and Vascular Biology 2007; 27:1866-1868
  18. Lohse P, Maas S, Lohse P, Elleder M, Kirk JM, Besley GTN, Seidel D. Compound heterozygosity for a Wolman mutation is frequent among patients with cholesteryl ester storage disease. Journal of Lipid Research 2000; 41:23-31
  19. Reiner Ž, Guardamagna O, Nair D, Soran H, Hovingh K, Bertolini S, Jones S, Ćorić M, Calandra S, Hamilton J, Eagleton T, Ros E. Lysosomal acid lipase deficiency – An under-recognized cause of dyslipidaemia and liver dysfunction. Atherosclerosis 2014; 235:21-30
  20. Valles-Ayoub Y, Esfandiarifard S, No D, Sinai P, Khokher Z, Kohan M, Kahen T, Darvish D. Wolman Disease (LIPA p.G87V) Genotype Frequency in People of Iranian-Jewish Ancestry. Genetic Testing and Molecular Biomarkers 2011; 15:395-398
  21. Reynolds TM, Mewies C, Hamilton J, Wierzbicki AS, group PPC. Identification of rare diseases by screening a population selected on the basis of routine pathology results-the PATHFINDER project: lysosomal acid lipase/cholesteryl ester storage disease substudy. J Clin Pathol 2018; 71:608-613
  22. Wolman M, Sterk VV, Gatt S, Frenkel M. PRIMARY FAMILIAL XANTHOMATOSIS WITH INVOLVEMENT AND CALCIFICATION OF THE ADRENALS. Pediatrics 1961; 28:742-757
  23. Jones SA, Valayannopoulos V, Schneider E, Eckert S, Banikazemi M, Bialer M, Cederbaum S, Chan A, Dhawan A, Di Rocco M, Domm J, Enns GM, Finegold D, Gargus JJ, Guardamagna O, Hendriksz C, Mahmoud IG, Raiman J, Selim LA, Whitley CB, Zaki O, Quinn AG. Rapid progression and mortality of lysosomal acid lipase deficiency presenting in infants. Genet Med 2016; 18:452-458
  24. Zhang B, Porto AF. Cholesteryl Ester Storage Disease. Journal of Pediatric Gastroenterology &amp; Nutrition2013; 56:682-685
  25. Tebani A, Sudrie-Arnaud B, Boudabous H, Brassier A, Anty R, Snanoudj S, Abergel A, Abi Warde MT, Bardou-Jacquet E, Belbouab R, Blanchet E, Borderon C, Bronowicki JP, Cariou B, Carette C, Dabbas M, Dranguet H, de Ledinghen V, Ferrieres J, Guillaume M, Krempf M, Lacaille F, Larrey D, Leroy V, Musikas M, Nguyen-Khac E, Ouzan D, Perarnau JM, Pilon C, Ratzlu V, Thebaut A, Thevenot T, Tragin I, Triolo V, Verges B, Vergnaud S, Bekri S. Large-scale screening of lipase acid deficiency in at risk population. Clin Chim Acta 2021; 519:64-69
  26. Pericleous M, Kelly C, Wang T, Livingstone C, Ala A. Wolman's disease and cholesteryl ester storage disorder: the phenotypic spectrum of lysosomal acid lipase deficiency. Lancet Gastroenterol Hepatol 2017; 2:670-679
  27. Maciejko JJ. Managing Cardiovascular Risk in Lysosomal Acid Lipase Deficiency. Am J Cardiovasc Drugs2017; 17:217-231
  28. Elleder M, Chlumska A, Hyanek J, Poupetova H, Ledvinova J, Maas S, Lohse P. Subclinical course of cholesteryl ester storage disease in an adult with hypercholesterolemia, accelerated atherosclerosis, and liver cancer. J Hepatol 2000; 32:528-534
  29. Wilson DP, Friedman M, Marulkar S, Hamby T, Bruckert E. Sebelipase alfa improves atherogenic biomarkers in adults and children with lysosomal acid lipase deficiency. J Clin Lipidol 2018; 12:604-614
  30. Burton BK, Deegan PB, Enns GM, Guardamagna O, Horslen S, Hovingh GK, Lobritto SJ, Malinova V, McLin VA, Raiman J, Di Rocco M, Santra S, Sharma R, Sykut-Cegielska J, Whitley CB, Eckert S, Valayannopoulos V, Quinn AG. Clinical Features of Lysosomal Acid Lipase Deficiency. J Pediatr Gastroenterol Nutr 2015; 61:619-625
  31. Kostner GM, Hadorn B, Roscher A, Zechner R. Plasma lipids and lipoproteins of a patient with cholesteryl ester storage disease. Journal of Inherited Metabolic Disease 1984; 8:9-12
  32. Ambler GK, Hoare M, Brais R, Shaw A, Butler A, Flynn P, Deegan P, Griffiths WJH. Orthotopic liver transplantation in an adult with cholesterol ester storage disease. JIMD Rep 2013; 8:41-46
  33. Hůlková H, Elleder M. Distinctive histopathological features that support a diagnosis of cholesterol ester storage disease in liver biopsy specimens. Histopathology 2012; 60:1107-1113
  34. Hamilton J, Jones I, Srivastava R, Galloway P. A new method for the measurement of lysosomal acid lipase in dried blood spots using the inhibitor Lalistat 2. Clinica Chimica Acta 2012; 413:1207-1210
  35. Bays H, Cohen DE, Chalasani N, Harrison Stephen A. An assessment by the Statin Liver Safety Task Force: 2014 update. Journal of Clinical Lipidology 2014; 8:S47-S57
  36. Ginsberg HN, Le NA, Short MP, Ramakrishnan R, Desnick RJ. Suppression of apolipoprotein B production during treatment of cholesteryl ester storage disease with lovastatin. Implications for regulation of apolipoprotein B synthesis. J Clin Invest 1987; 80:1692-1697
  37. Di Bisceglie AM, Ishak KG, Rabin L, Hoeg JM. Cholesteryl ester storage disease: Hepatopathology and effects of therapy with lovastatin. Hepatology 1990; 11:764-772
  38. Balwani M, Breen C, Enns GM, Deegan PB, Honzík T, Jones S, Kane JP, Malinova V, Sharma R, Stock EO, Valayannopoulos V, Wraith JE, Burg J, Eckert S, Schneider E, Quinn AG. Clinical effect and safety profile of recombinant human lysosomal acid lipase in patients with cholesteryl ester storage disease. Hepatology 2013; 58:950-957
  39. Burton BK, Balwani M, Feillet F, Barić I, Burrow TA, Camarena Grande C, Coker M, Consuelo-Sánchez A, Deegan P, Di Rocco M, Enns GM, Erbe R, Ezgu F, Ficicioglu C, Furuya KN, Kane J, Laukaitis C, Mengel E, Neilan EG, Nightingale S, Peters H, Scarpa M, Schwab KO, Smolka V, Valayannopoulos V, Wood M, Goodman Z, Yang Y, Eckert S, Rojas-Caro S, Quinn AG. A Phase 3 Trial of Sebelipase Alfa in Lysosomal Acid Lipase Deficiency. New England Journal of Medicine 2015; 373:1010-1020
  40. Jones SA, Rojas-Caro S, Quinn AG, Friedman M, Marulkar S, Ezgu F, Zaki O, Gargus JJ, Hughes J, Plantaz D, Vara R, Eckert S, Arnoux JB, Brassier A, Le Quan Sang KH, Valayannopoulos V. Survival in infants treated with sebelipase Alfa for lysosomal acid lipase deficiency: an open-label, multicenter, dose-escalation study. Orphanet J Rare Dis 2017; 12:25
  41. Valayannopoulos V, Malinova V, Honzík T, Balwani M, Breen C, Deegan PB, Enns GM, Jones SA, Kane JP, Stock EO, Tripuraneni R, Eckert S, Schneider E, Hamilton G, Middleton MS, Sirlin C, Kessler B, Bourdon C, Boyadjiev SA, Sharma R, Twelves C, Whitley CB, Quinn AG. Sebelipase alfa over 52 weeks reduces serum transaminases, liver volume and improves serum lipids in patients with lysosomal acid lipase deficiency. Journal of hepatology 2014; 61:1135-1142
  42. Vijay S, Brassier A, Ghosh A, Fecarotta S, Abel F, Marulkar S, Jones SA. Long-term survival with sebelipase alfa enzyme replacement therapy in infants with rapidly progressive lysosomal acid lipase deficiency: final results from 2 open-label studies. Orphanet J Rare Dis 2021; 16:13
  43. Burton BK, Feillet F, Furuya KN, Marulkar S, Balwani M. Sebelipase alfa in children and adults with lysosomal acid lipase deficiency: Final results of the ARISE study. J Hepatol 2022; 76:577-587
  44. Kohli R, Ratziu V, Fiel MI, Waldmann E, Wilson DP, Balwani M. Initial assessment and ongoing monitoring of lysosomal acid lipase deficiency in children and adults: Consensus recommendations from an international collaborative working group. Molecular Genetics and Metabolism 2020; 129:59-66

Anatomy and Ultrastructure of Bone – Histogenesis, Growth and Remodeling

ABSTRACT

 

Bones have three major functions: to serve as mechanical support, sites of muscle insertion and as a reserve of calcium and phosphate for the organism. Recently, a fourth function has been attributed to the skeleton: an endocrine organ. The organic matrix of bone is formed mostly of collagen, but also non-collagenous proteins. Hydroxyapatite crystals bind to both types of proteins. Most components of the bone matrix are synthesized and secreted by osteoblasts.  Resorption of the bone matrix is required for adaptation to growth, repair and mineral mobilization. This process is performed by the macrophage-related osteoclast. Bone is remodeled throughout life through a coordinated sequence of events which involve the sequential actions of osteoclasts and osteoblasts, replacing old bone with new bone. In the normal adult skeleton, remodeling is coupled such that the level of resorption is equal to the level of formation and bone density remains constant. Intramembranous ossification is the process by which flat bones are formed. For this process, osteoblasts differentiate directly from mesenchymal cells to form the bone matrix. Long bones are formed by endochondral ossification, which is characterized by the presence of a cartilaginous model in which chondrocytes differentiate and mineralized cartilage is replaced with bone through remodeling.

 

INTRODUCTION

 

Bone, a specialized and mineralized connective tissue, makes up, with cartilage, the skeletal system, which serves three main functions: A mechanical function as support and site of muscle attachment for locomotion; a protective function for vital organs and bone marrow; and finally a metabolic function as a reserve of calcium and phosphate used for the maintenance of serum homeostasis, which is essential to life.  Recently, a fourth important function has been attributed to bone tissue – that of an endocrine organ.  Bone cells produce fibroblast growth factor 23 (FGF23) and osteocalcin. FGF23 regulates phosphate handling in the kidney and osteocalcin regulates energy and glucose metabolism (see below) (1,2).

 

In this chapter the anatomy and cell biology of bone is described as well as the mechanisms of bone remodeling, development, and growth. Remodeling is the process by which bone is turned-over, allowing the maintenance of the shape, quality, and amount of the skeleton. This process is characterized by the coordinated actions of osteoclasts and osteoblasts, organized in bone multicellular units (BMUs) which follow an Activation-Resorption-Formation sequence of events. During embryonic development, bone formation occurs by two different means: intramembranous ossification and endochondral ossification. Bone Growth is a term used to describe the changes in bone structure once the skeleton is formed and during the period of skeletal growth and maturation.

 

BONE AS AN ORGAN: MACROSCOPIC ORGANIZATION

 

Two types of bones are found in the skeleton: flat bones (skull bones, scapula, mandible, and ileum) and long bones (tibia, femur, humerus, etc.). These are derived by two distinct types of development: intramembranous and endochondral, respectively, although the development and growth of long bones actually involve both cellular processes. The main difference between intramembranous and endochondral bone formation is the presence of a cartilaginous model, or anlage, in the latter.

Long bones have two wider extremities (the epiphyses), a cylindrical hollow portion in the middle (the midshaft or diaphysis), and a transition zone between them (the metaphysis). The epiphysis on the one hand and the metaphysis and midshaft on the other hand originate from two independent ossification centers, and are separated by a layer of cartilage, the epiphyseal cartilage (which also constitutes the growth plate) during the period of development and growth. This layer of proliferative cells and expanding cartilage matrix is responsible for the longitudinal growth of bones; it progressively mineralizes and is later remodeled and replaced by bone tissue by the end of the growth period (see section on Skeletal Development). The external part of the bones is formed by a thick and dense layer of calcified tissue, the cortex (compact bone) which, in the diaphysis, encloses the medullary cavity where the hematopoietic bone marrow is housed. Toward the metaphysis and the epiphysis, the cortex becomes progressively thinner and the internal space is filled with a network of thin, calcified trabeculae forming the cancellous or trabecular bone. The spaces enclosed by these thin trabeculae are also filled with hematopoietic bone marrow and are continuous with the diaphyseal medullary cavity. The outer cortical bone surfaces at the epiphyses are covered with a layer of articular cartilage that does not calcify.

 

Bone is consequently in contact with the soft tissues along two surfaces: an external surface (the periosteal surface) and an internal surface (the endosteal surface). These surfaces are lined with osteogenic cells along the periosteum and the endosteum, respectively.

 

Cortical and trabecular bone are made up of the same cells and the same matrix elements, but there are structural and functional differences. The primary structural difference is quantitative: 80% to 90% of the volume of compact bone is calcified, whereas only 15% to 25% of the trabecular volume is calcified (the remainder being occupied by bone marrow, blood vessels, and connective tissue). The result is that 70% to 85% of the interface with soft tissues is at the endosteal bone surface, including all trabecular surfaces, leading to the functional difference: the cortical bone fulfills mainly a mechanical and protective function and the trabecular bone mainly a metabolic function, albeit trabeculae definitively participate in the biomechanical function of bones, particularly in bones like the vertebrae.

 

Recently, more attention has been given to cortical bone structure since cortical porosity is intimately linked to the remodeling process as well as to bone strength.  Indeed, an increase in cortical porosity is associated with an increase in fragility fractures (3).

 

BONE AS A TISSUE: BONE MATRIX AND MINERAL

 

Bone matrix consists mainly of type I collagen fibers (approximately 90%) and non-collagenous proteins. Within lamellar bone, the fibers are forming arches for optimal bone strength. This fiber organization allows the highest density of collagen per unit volume of tissue. The lamellae can be parallel to each other if deposited along a flat surface (trabecular bone and periosteum), or concentric if deposited on a surface surrounding a channel centered on a blood vessel (cortical bone Haversian system). Spindle- or plate-shaped crystals of hydroxyapatite [3Ca 3 (PO 42 ·(OH) 2] are found on the collagen fibers, within them, and in the matrix around. They tend to be oriented in the same direction as the collagen fibers.

When bone is formed very rapidly during development and fracture healing, or in tumors and some metabolic bone diseases, there is no preferential organization of the collagen fibers. They are then not as tightly packed and found in somewhat randomly oriented bundles: this type of bone is called woven bone, as opposed to lamellar bone. Woven bone is characterized by irregular bundles of collagen fibers, large and numerous osteocytes, and delayed, disorderly calcification which occurs in irregularly distributed patches. Woven bone is progressively replaced by mature lamellar bone during the remodeling process that follows normally development or healing (see below).

 

Numerous non-collagenous proteins present in bone matrix have been purified and sequenced, but their role has been only partially characterized (Table 1) (4). Most non-collagenous proteins within the bone matrix are synthesized by osteoblasts, but not all: approximately a quarter of the bone non-collagenous proteins are plasma proteins which are preferentially absorbed by the bone matrix, such as a 2-HS-glycoprotein, which is synthesized in the liver. The major non-collagenous protein produced is osteocalcin, which makes up 1% of the matrix, and may play a role in calcium binding and stabilization of hydroxyapatite in the matrix and/or regulation of bone formation, as suggested by increased bone mass in osteocalcin knockout mice. Another negative regulator of bone formation found in the matrix is matrix gla protein, which appears to inhibit premature or inappropriate mineralization, as demonstrated in a knockout mouse model. In contrast to this, biglycan, a proteoglycan, is expressed in the bone matrix, and positively regulates bone formation, as demonstrated by reduced bone formation and bone mass in biglycan knockout mice.  Osteocalcin has recently been shown to have an important endocrine function acting on the pancreatic beta cell.  Its hormonally active form (undercarboxylated osteocalcin, stimulates insulin secretion and enhances insulin sensitivity in adipose tissues and muscle, improving glucose utilization in peripheral tissues (2).

 

Table 1. Non-Collagenous Proteins in Bone (4)

PROTEIN

MW

ROLE

Osteonectin (SPARC)

32K

Calcium, apatite and matrix protein binding

Modulates cell attachment

α-2-HS-Glycoprotein

46-67K

Chemotactic for monocytes

Mineralization via matrix vesicles

Osteocalcin (Bone GLA protein)

6K

Involved in stabilization of hydroxyapatite

Binding of calcium

Chemotactic for monocytes

Regulation of bone formation

Matrix-GLA-protein

9K

Inhibits matrix mineralization

Osteopontin

(Bone Sialoprotein I)

50K

Cell attachment (via RGD sequence)

Calcium binding

Bone Sialoprotein II

75K

Cell attachment (via RGD sequence)

Calcium binding

24K Phosphoprotein

(α-1(I) procollagen N-propeptide)

24K

Residue from collagen processing

Biglycan (Proteoglycan I)

45K core

Regulation of collagen fiber growth

Mineralization and bone formation

Growth factor binding

Decorin (Proteoglycan II)

36K core + side chains

Collagen fibrillogenesis

Growth factor binding

Thrombospondin & Fibronectin

 

Cell attachment (via RGD sequence)

Growth factor binding

Hydroxyapatite formation

Others (including proteolipids

 

Mineralization

Growth Factors

IGFI & IGFII

TGFβ

Bone morphogenetic proteins (BMPs)

 

Differentiation, proliferation and activity of osteoblasts

Induction of bone and cartilage in osteogenesis and fracture repair

 

CELLULAR ORGANIZATION WITHIN THE BONE MATRIX: OSTEOCYTES

 

The calcified bone matrix is not metabolically inert, and cells (osteocytes) are found embedded deep within the bone in small lacunae (Figure 1). All osteocytes are derived from bone forming cells (osteoblasts) which have been trapped in the bone matrix that they produced and which became calcified. Even though the metabolic activity of the osteoblast decreases dramatically once it is fully encased in bone matrix, now becoming an osteocyte, these cells still produce matrix proteins.

 

Figure 1. Wnt signaling determines the cell fate of mesenchymal progenitor cells and regulates bone formation and resorption. The Wnt canonical pathway represses adipocyte differentiation and chondrocyte differentiation from progenitor cells, whereas it is required for the transition of chondrocytes to hypertrophy. In contrast, Wnt pathway activation promotes the osteoblast cell lineage by controlling proliferation, maturation, terminal differentiation, and bone formation. Differentiated osteoblasts and/or osteocytes produce Wnt inhibitors such as Dickkopf (Dkk1) and sclerostin (Sost) proteins as a negative feedback control of osteoblast differentiation and function. Wnt signaling also induces osteoblasts to produce more osteoprotegerin (OPG), increasing the ratio of OPG to receptor activator of NF-κB ligand (RANKL) to decrease osteoclast differentiation and bone resorption.

 

Osteocyte morphology varies according to cell age and functional activity. A young osteocyte has most of the ultrastructural characteristics of the osteoblast from which it was derived, except that there has been a decrease in cell volume and in the importance of the organelles involved in protein synthesis (rough endoplasmic reticulum, Golgi). An older osteocyte, located deeper within the calcified bone, shows a further decrease in cell volume and organelles, and an accumulation of glycogen in the cytoplasm. These cells synthesize small amounts of new bone matrix at the surface of the osteocytic lacunae, which can subsequently calcify. Osteocytes express, in low levels, a number of osteoblast markers, including osteocalcin, osteopontin, osteonectin and the osteocyte marker E11.

 

Osteocytes have numerous long cell processes rich in microfilaments, which are in contact with cell processes from other osteocytes (there are frequent gap junctions), or with processes from the cells lining the bone surface (osteoblasts or flat lining cells). These processes are organized during the formation of the matrix and before its calcification; they form a network of thin canaliculi permeating the entire bone matrix. Osteocytic canaliculi are not distributed evenly around the cell, but are mainly directed toward the bone surface. Between the osteocyte's plasma membrane and the bone matrix itself is the periosteocytic space. This space exists both in the lacunae and in the canaliculi, and it is filled with extracellular fluid (ECF), the only source of nutrients, cytokines and hormones for the osteocyte. ECF flow through the canalicular network is altered during bone matrix compression and tension and is believed not only to allow exchanges with the extracellular fluids in the surrounding tissues but also to create shear forces that are directly involved in mechanosensing and regulation of bone remodeling. Current understanding of mechanotransduction is based upon the presence of a mechanosensing cilium at the level of the osteocyte’s cell body, capable of detecting the changes in fluid flow determined by mechanical loading of bone. In turn, the activation of the mechanosensing cilium may determine the local concentration of cytokines capable of regulating bone formation and/or bone resorption, such as RANKL, OPG or sclerostin (see below).

 

Indeed, given the structure of the network and the location of osteocytes within lacunae where ECF flow can be detected, it is likely that osteocytes respond to bone tissue strain and influence bone remodeling activity by recruiting osteoclasts to sites where bone remodeling is required. Osteocyte cellular activity is increased after bone loading; studies in cell culture have demonstrated increased calcium influx and prostaglandin production by osteocytes after mechanical stimulation, but there is no direct evidence for osteocytes signaling to cells on the bone surface in response to bone strain or microdamage to date. Osteocytes can become apoptotic and their programmed cell death may be one of the critical signals for induction of bone remodeling. Ultimately, the fate of the osteocyte is to be phagocytosed and digested together with the other components of bone during osteoclastic bone resorption. The recent ability to isolate and culture osteocytes, as well as the creation of immortalized osteocytic cell lines now allows the study of these cells at the molecular level and this is expected to significantly further our understanding of their role in bone biology and disease.(5) In particular, the discoveries that osteocytes can secrete the Wnt antagonist sclerostin and that this secretion is inhibited both by PTH treatment and by mechanical loading establishes the first direct link between biomechanics, endocrine hormones, bone formation and osteocytes. Similarly, osteocytes can secrete RANKL and OPG, contributing also to the regulation of bone resorption. Thus, osteocytes are emerging as the critical cell type linking mechanical forces in bone to the regulation of bone mass and shape through remodeling.

 

THE OSTEOBLAST AND BONE FORMATION  

 

The osteoblast is the bone lining cell responsible for the production of the bone matrix constituents, collagen and non-collagenous proteins (Figure 2). Osteoblasts never appear or function individually but are always found in clusters of cuboidal cells along the bone surface (~100–400 cells per bone-forming site).

Figure 2. Osteocyte. Electron micrograph of an osteocyte within a lacuna in calcified bone matrix. The cell has a basal nucleus, cytoplasmic extensions, and well-developed Golgi and endoplasmic reticulum.

 

Osteoblasts do not operate in isolation and gap junctions are often found between osteoblasts working together on the bone surface. Osteoblasts also appear to communicate with the osteocyte network within the bone matrix (see above), since cytoplasmic processes on the secreting side of the osteoblast extend deep into the osteoid matrix and are in contact with processes of the osteocytes dwelling there.

 

At the light microscope level, the osteoblast is characterized morphologically by a round nucleus at the base of the cell (away from the bone surface), an intensely basophilic cytoplasm, and a prominent Golgi complex located between the nucleus and the apex of the cell. Osteoblasts are always found lining the layer of bone matrix that they are producing, but before it is calcified (osteoid tissue). Osteoid tissue exists because of a time lag of approximately 10 days between matrix formation and its subsequent calcification. Behind the osteoblast can usually be found one or two layers of cells: activated mesenchymal cells and preosteoblasts (see below). A mature osteoblast does not divide.

 

At the ultrastructural level, the osteoblast is characterized by the presence of a well-developed rough endoplasmic reticulum with dilated cisternae and a dense granular content, and the presence of a large circular Golgi complex comprising multiple Golgi stacks. These organelles are involved in the major activity of the osteoblast: the production and secretion of collagenous and non-collagenous bone matrix proteins, including type I collagen. Osteoblasts also produce a range of growth factors under a variety of stimuli, including the insulin-like growth factors (IGFs), platelet-derived growth factors (PDGFs), basic fibroblast growth factor (bFGF), transforming growth factor-beta (TGFb), a range of cytokines, the bone morphogenetic proteins (BMPs and Wnts.(3) Osteoblast activity is regulated in an autocrine and paracrine manner by these growth factors, whose receptors can be found on osteoblasts, as well as receptors for a range of endocrine hormones. Classic endocrine receptors include receptors for parathyroid hormone/ parathyroid hormone related protein receptor, thyroid hormone, growth hormone, insulin, progesterone and prolactin. Osteoblastic nuclear steroid hormone receptors include receptors for estrogens, androgens, vitamin D 3 and retinoids. Receptors for paracrine and autocrine effectors include those for epidermal growth factor (EGF), IGFs, PDGF, TGFb, interleukins, FGFs, BMPs and Wnts (LRP5/6 and Frizzled) (6,7) Osteoblasts also have receptors for several adhesion molecules (integrins) involved in cell attachment to the bone surface.

 

Among the cytokines secreted by the osteoblast are the main regulators of osteoclast differentiation, i.e. M-CSF, RANKL and osteoprotegerin (OPG) (8,9). M-CSF is essential in inducing the commitment of monocytes to the osteoclast lineage whereas RANKL promotes the differentiation and activity of osteoclasts (see below).

 

Osteoblasts originate from local pluripotent mesenchymal stem cells, either bone marrow stromal stem cells (endosteum) or connective tissue mesenchymal stem cells (periosteum). These precursors, with the right stimulation, undergo proliferation and differentiate into preosteoblasts, at which point they are committed to differentiate into mature osteoblasts.

 

The committed preosteoblast is located in apposition to the bone surface, and usually present in layers below active mature osteoblasts. They are elliptical cells, with an elongated nucleus, and are still capable of proliferation. Preosteoblasts lack the well-developed protein synthesizing capability of the mature osteoblast, and do not have the characteristically localized, mature rough endoplasmic reticulum or Golgi apparatus of the mature cell.

 

The development of the osteoblast phenotype is gradual, with a defined sequence of gene expression and cell activity during development and maturation, controlled by a sequence of transcription factors and cytokines (Figure 3).

Figure 3. Osteoblasts and Osteoid Tissue. A: Light micrograph of a group of osteoblasts producing osteoid; note the newly embedded osteocyte. B: Electron micrograph of 3 osteoblasts covering a layer of mineralizing osteoid tissue. Note the prominent Golgi and endoplasmic reticulum characteristic of active osteoblasts. The black clusters in the osteoid tissue are deposits of mineral. C: Osteoblast Lineage. Osteoblasts originate from undifferentiated mesenchymal cells which are capable of proliferation and which may differentiate into one of a range of cell types. The preosteoblast is also capable of proliferation and may be already committed to an osteoblast phenotype. The mature osteoblast no longer proliferates, but can differentiate further into an osteocyte once embedded in the bone matrix, or to a lining cell on the bone surface.

 

Two transcription factors, Runx2 and Osterix (Osx), which is downstream of Runx2, are absolutely required for osteoblast differentiation. Runx2 is expressed in mesenchymal condensations and chondrocytes, in addition to osteoblasts. Runx2 target genes include several genes expressed by the mature osteoblast including osteocalcin, bone sialoprotein, osteopontin and collagen a1(I), as well as the Runx2 gene itself. Osx may be mostly important for pushing precursors cells away from the chondrocyte and into the osteoblast lineage.

 

The most important breakthrough in the understanding of the regulation of bone formation in recent years, is the finding of a clear link between LRP5, a co-receptor for Wnts, and bone mass in humans and in mice. Loss of function in LRP5 leads to the Osteoporosis Pseudo-Glioma syndrome (OPPG), with extremely low bone mass, whereas gain of function leads to the High Bone Mass (HBM) phenotype in humans. In addition, deletion mutations in the gene encoding sclerostin (Sost), another endogenous inhibitor of the Wnt pathway, also lead to osteosclerotic phenotypes (Sclerosteosis, Van Buchem syndrome).(7) These findings have opened a whole new field of investigation both in terms of understanding the mechanism that regulate osteoblasts and their bone-matrix secreting activity and in terms of drug discovery in the hope to target one component of the Wnt signaling pathway and thereby increase bone mass in osteoporotic patients. Of note, in 2019, the FDA approved romosozumab, a monoclonal antibody to sclerostin, for the treatment of postmenopausal women with osteoporosis at high risk for fracture.

 

Toward the end of the matrix secreting period, a further step is involved in osteoblast maturation. Approximately 15% of the mature osteoblasts become encapsulated in the new bone matrix and differentiate into osteocytes. In contrast, some cells remain on the bone surface, becoming flat lining cells.

 

Mechanism of Bone Formation

 

Bone formation occurs by three coordinated processes: the production of osteoid matrix, its maturation, and the subsequent mineralization of the matrix. In normal adult bone, these processes occur at the same rate, so that the balance between matrix production and mineralization is equal. Initially, osteoblasts deposit collagen rapidly, without mineralization, producing a thickening osteoid seam. This is followed by an increase in the mineralization rate to equal the rate of collagen synthesis. In the final stage, the rate of collagen synthesis decreases, and mineralization continues until the osteoid seam is fully mineralized. This time lag (termed the mineralization lag time or osteoid maturation period) appears to be required for osteoid to be modified so it is able to support mineralization. While this delay is not yet understood, it is likely that either collagen cross-linking occurs or an inhibitor of mineralization, such as matrix gla protein, is removed during this time, thus allowing mineralization to proceed.

 

To initiate mineralization in woven bone, or in growth plate cartilage, high local concentrations of Ca2+ and PO43- ions must be reached in order to induce their precipitation into amorphous calcium phosphate, leading to hydroxyapatite crystal formation. This is achieved by membrane-bound matrix vesicles, which originate by budding from the cytoplasmic processes of the chondrocyte or the osteoblast and are deposited within the matrix during its formation. In the matrix, these vesicles are the first structure wherein hydroxyapatite crystals are observed. The membranes are very rich in alkaline phosphatases and in acidic phospholipids, which hydrolyze inhibitors of calcification in the matrix including pyrophosphate and ATP allowing condensation of apatite crystals. Once the crystals are in the matrix environment, they will grow in clusters which later coalesce to completely calcify the matrix, filling the spaces between and within the collagen fibers. In adult lamellar bone, matrix vesicles are not present, and mineralization occurs in an orderly manner through progression of the mineralization front into the osteoid tissue.

 

THE OSTEOCLAST AND BONE RESORPTION

 

The osteoclast is the bone lining cell responsible for bone resorption (Figure 4). The osteoclast is a giant multinucleated cell, up to 100mm in diameter and containing four to 20 nuclei. It is usually found in contact with a calcified bone surface and within a lacuna (Howship's lacunae) that is the result of its own resorptive activity. It is possible to find up to four or five osteoclasts in the same resorptive site, but there are usually only one or two. Under the light microscope, the nuclei appear to vary within the same cell: some are round and euchromatic, and some are irregular in contour and heterochromatic, possibly reflecting asynchronous fusion of mononuclear precursors. The cytoplasm is "foamy" with many vacuoles. The zone of contact with the bone is characterized by the presence of a ruffled border with dense patches on each side (the sealing zone).

Figure 4. Osteoclasts and the Mechanism of Bone Resorption. A: Light micrograph and B: electron micrograph of an osteoclast, demonstrating the ruffled border and numerous nuclei. C: Osteoclastic resorption. The osteoclast forms a sealing zone via integrin mediated attachment to specific peptide sequences within the bone matrix, forming a sealed compartment between the cell and the bone surface. This compartment is acidified such that an optimal pH is reached for lysosomal enzyme activity and bone resorption.

 

Characteristic ultrastructural features of this cell are abundant Golgi complexes around each nucleus, mitochondria, and transport vesicles loaded with lysosomal enzymes. The most prominent features of the osteoclast are, however, the deep foldings of the plasma membrane in the area facing the bone matrix (ruffled border) and the surrounding zone of attachment (sealing zone). The sealing zone is formed by a ring of focal points of adhesion (podosomes) with a core of actin and several cytoskeletal and regulatory proteins around it, that attach the cell to the bone surface, thus sealing off the subosteoclastic bone-resorbing compartment. The attachment of the cell to the matrix is performed via integrin receptors, which bind to specific RGD (Arginine-Glycine-Aspartate) sequences found in matrix proteins (see Table 1). The plasma membrane in the ruffled border area contains proteins that are also found at the limiting membrane of lysosomes and related organelles, and a specific type of electrogenic vacuolar proton ATPase involved in acidification. The basolateral plasma membrane of the osteoclast is specifically enriched in Na+, K+-ATPase (sodium pumps), HCO 3 - /Cl -exchangers, and Na+/H+ exchangers and numerous ion channels (10).

 

Lysosomal enzymes such as tartrate resistant acid phosphatase and cathepsin K are actively synthesized by the osteoclast and are found in the endoplasmic reticulum, Golgi, and many transport vesicles. The enzymes are secreted, via the ruffled border, into the extracellular bone-resorbing compartment where they reach a sufficiently high extracellular concentration because this compartment is sealed off. The transport and targeting of these enzymes for secretion at the apical pole of the osteoclast involves mannose-6-phosphate receptors. Furthermore, the cell secretes several metalloproteinases such as collagenase (MMP-13) and gelatinase B (MMP-9) which appear to be involved in preosteoclast migration to the bone surface as well as bone matrix digestion. Among the key enzymes being synthesized and secreted by the osteoclast is cathepsin K, an enzyme capable or degrading collagen at low pH and a target for inhibition of bone resorption. (11)

 

Attachment of the osteoclast to the bone surface is essential for bone resorption. This process involves transmembrane adhesion receptors of the integrin. Integrins attach to specific amino acid sequences (mostly RGD sequences) within proteins in or at the surface of the bone matrix. In the osteoclast, avb3 (vitronectin receptor), a2b1 (collagen receptor) and avb5 integrins are predominantly expressed. Without cell attachment the acidified microenvironment cannot be established and the osteoclast cannot be highly mobile, a functional property associated with the formation of podosomes.

 

After osteoclast adhesion to the bone matrix, avb3 binding activates cytoskeletal reorganization within the osteoclast, including cell spreading and polarization. In most cells, cell attachment occurs via focal adhesions, where stress fibers (bundles of microfilaments) anchor the cell to the substrate. In osteoclasts, attachment occurs via podosomes. Podosomes are more dynamic structures than focal adhesions, and occur in cells that are highly motile. It is the continual assembly and disassembly of podosomes that allows osteoclast movement across the bone surface during bone resorption. Integrin signaling and subsequent podosome formation is dependent on a number of adhesion kinases including the proto-oncogene src, which, while not required for osteoclast maturation, is required for osteoclast function, as demonstrated by osteopetrosis in the src knockout mouse. Pyk2, another member of the focal adhesion kinase family is also activated by avb3 during osteoclast attachment, and is required for bone resorption.(10) Several actin-regulatory proteins have also been shown to be present in podosomes and required for bone resorption, again pointing to the importance of integrin signaling and podosome assembly and disassembly in the function of osteoclasts. (12)

 

Osteoclasts resorb bone by acidification and proteolysis of the bone matrix and hydroxyapatite crystals encapsulated within the sealing zone. Carbonic anhydrase type II produces hydrogen ions within the cell, which are then pumped across the ruffled border membrane via proton pumps located in the basolateral membrane, thereby acidifying the extracellular compartment. The protons are highly concentrated in the cytosol of the osteoclast; ATP and CO2 are provided by the mitochondria. The basolateral membrane activity exchanges bicarbonate for chloride, thereby avoiding alkalization of the cytosol. K+ channels in the basolateral domain and Cl - channels in the apical ruffled border ensure dissipation of the electrogenic gradients generated by the vacuolar H+-ATPase The basolateral sodium pumps might be involved in secondary active transport of calcium and/or protons in association with a Na + /Ca 2+ exchanger and/or a Na+/H+ antiport. Genetic mutations in several of these components of the acidification and ion transport systems have been shown to be associated with osteopetrosis (defective bone resorption by osteoclasts) in humans and in mice.

 

The first process during bone matrix resorption is mobilization of the hydroxyapatite crystals by digestion of their link to collagen via the non-collagenous proteins and the low pH dissolves the hydroxyapatite crystals, exposing the bone matrix. Then the residual collagen fibers are digested by cathepsin K, now at optimal pH. The residues from this extracellular digestion are either internalized, or transported across the cell and released at the basolateral domain. Residues may also be released during periods of sealing zone relapse, as probably occurs during osteoclast motility, and possibly induced by a calcium sensor responding to the rise of extracellular calcium in the bone-resorbing compartment.

 

The regulation of bone resorption is mostly mediated by the action of hormones on stromal cells, osteoblasts and osteocytes. For example, PTH can stimulate osteoblastic production of M-CSF, RANKL, OPG or IL-6, which then act directly on the osteoclast (5,6).

 

Origin and Fate of the Osteoclast (6)

 

The osteoclast derives from cells in the mononuclear phagocyte lineage (Figure 5). Their differentiation requires the transcription factors PU-1 and MiTf at early stages, committing the precursors into the myeloid lineage. M-CSF is then required to engage the cells in the monocyte lineage and ensure their proliferation and the expression of the RANK receptor. At that stage, the cells require the presence of RANKL, a member of the TNF family of cytokines produced by stromal cells, to truly commit to the osteoclast lineage and progress in their differentiation program. This step also requires expression of TRAF6, NFκB, c-Fos and NFAT c1, all downstream effectors of RANK signaling. Although this differentiation occurs at the early promonocyte stage, monocytes and macrophages already committed to their own lineage might still be able to form osteoclasts under the right stimuli. Despite its mononuclear phagocytic origin, the osteoclast membrane express distinct markers: it is devoid of Fc and C 3 receptors, as well as of several other macrophage markers; like mononuclear phagocytes, however, the osteoclast is rich in nonspecific esterases, synthesizes lysozyme, and expresses CSF-1 receptors. Monoclonal antibodies have been produced that recognize osteoclasts but not macrophages. The osteoclast, unlike macrophages, also expresses, millions of copies of the RANK, calcitonin, and vitronectin (integrin avb3) receptors. Whether it expresses receptors for parathyroid hormone, estrogen, or vitamin D is still controversial. Dendritic cell-specific transmembrane protein (DC-STAMP) is currently considered to be the master regulator of osteoclastogenesis.  Knock out of DC-STAMP completely abrogates cell-cell fusion during osteoclastogenesis; osteoclasts isolated from DC-STAMP knock-out mice are mononucleated. (13) Another important factor involved in cell fusion is Pin 1, an enzyme that specifically recognizes the peptide bond between phosphorylated serine or threonine and proline.  Pin 1 regulates cell fusion during osteoclastogeneis by suppressing DC-STAMP. (14,15) Recent evidence suggest that the osteoclast undergoes apoptosis after a cycle of resorption, a process favored by estrogens, possibly explaining the increased bone resorption after gonadectomy or menopause.

Figure 5. Osteoclast Life Cycle. The osteoclast is derived from a mononuclear hematopoietic precursor cell which, upon activation, fuses with other precursors to form a multinucleated osteoclast. The osteoclast first attaches to the bone surface then commences resorption. After a cycle of bone resorption, the osteoclast undergoes apoptosis.

 

Relations to the Immune System (Osteoimmunology)

 

In the last few years it has been recognized that, in part due to the link between the osteoclast, macrophages and dendritic cells (all three belong to the same cell lineage), osteoclasts are regulated by and share regulatory mechanisms with cells of the immune system. For instance, T cells can produce locally RANKL, activating osteoclastogenesis. B cells may share a common precursor with and regulate osteoclast precursors. RANKL signaling and “immunoreceptor tyrosine-based activation motif” (ITAM) signals cooperate in osteoclastogenesis (16).

 

BONE REMODELING

 

Bone remodeling is the process by which bone is turned over; it is the result of the activity of the bone cells at the surfaces of bone, mainly the endosteal surface (which includes all trabecular surfaces). Remodeling is traditionally classified into two distinct types: Haversian remodeling within the cortical bone and endosteal remodeling along the trabecular bone surface. This distinction is more morphological than physiological because the Haversian surface is an extension of the endosteal surface and the cellular events during these two remodeling processes follow exactly the same sequence.

 

The Remodeling Sequence

 

Bone formation and bone resorption do not occur along the bone surface at random: they are coordinated as part of the turnover mechanism by which old bone is replaced by new bone, providing an opportunity to change the shape, architecture or density of the skeleton. In the normal adult skeleton, bone formation only occurs, for the most part, where bone resorption has already occurred. This basic principle of cellular activity at the remodeling site constitutes the Activation-Resorption-Reversal-Formation (ARRF) sequence (Figure 6).

Figure 6. The Bone Remodeling Sequence. The Activation-Resorption-Reversal-Formation cycle of bone remodeling as it occurs in trabecular bone. See text for details.

 

Under some signal, today considered to emanate from osteocytes, a locally acting factor released by lining cells, osteocytes, marrow cells, or in response to bone deformation or fatigue-related microfracture, a group of preosteoclasts are activated. These mononuclear cells attach to the bone via avb3 integrins and fuse to form a multi-nucleated osteoclast which will, in a definite area of the bone surface, resorb the bone matrix. After resorption of the bone, and osteoclast detachment, uncharacterized mononuclear cells cover the surface and a cement line is formed. The cement line marks the limit of bone resorption, and acts to cement together the old and the new bone. This is termed the reversal phase, and is followed by a period of bone formation. Preosteoblasts are activated, proliferate and differentiate into osteoblasts, which move onto the bone surface, forming an initial matrix (osteoid), which becomes mineralized after a time lag (the osteoid maturation period). The basic remodeling sequence is therefore Activation-Resorption-Formation; it is performed by a group of cells called the Basic Multicellular Unit (BMU). The complete remodeling cycle takes about 3 months in humans (Figure 7).

Figure 7. Bone Growth and Remodeling at the Growth Plate. The light micrograph demonstrates the zones of chondrocyte differentiation, as well as mineralization (black). The schematic representation shows the cellular events occurring at the growth plate in long bones. Note that bone formation in this process occurs by repeated Activation-Resorption-Formation cycles of bone remodeling beginning with the calcified cartilage matrix.

 

For decades, the reversal phase of the remodeling cycle was the least well understood.  It was recognized that during this phase, the resorption cavity was occupied by mononucleated cells, but the nature of these cells was unknown (17).  Recent work by Delaisse and colleagues (18) has definitively identified the reversal cells as belonging to the osteogenic lineage, expressing classic osteoblast markers: Runx2, ALP, and Col3. By applying immunocytochemistry and histomorphometry to femur and fibula samples harvested from teenagers and adults, these investigators have provided a much more complete picture of the temporal sequence of cellular events that occur between the start of resorption and the onset of formation.  In order to visualize the entire sequence of events, they analyzed longitudinal sections of evolving Haversian systems. They observed osteoclasts at two distinct locations: at the cutting cone (referred to as primary osteoclasts) and close to the reversal cells (referred to as secondary osteoclasts). The presence of secondary osteoclasts in the reversal phase suggests that bone resorption continues during this phase, which has been renamed the resorption-reversal phase. The authors have concluded that the primary osteoclasts are responsible for drilling the tunnel (initial resorption) and the secondary osteoclasts work to increase its diameter by radial resorption. This radial resorption was shown to be a major contributor to the overall amount of bone resorbed in each BMU. This new and more complete model of the resorption-reversal phase will lead to enhanced understanding of the delicate and all-important balance between resorption and formation (Figure 8).

 

Figure 8. Cartoon of a bone remodeling unit in cortical bone, showing the change in the designation of the reversal phase as a result of recent new findings. IR = initial resorption; RR = radial resorption; Og = osteoprogenitor cell; Oc = osteoclast. (17)

 

For many years it has been accepted that bone resorption and formation are coupled in the same way that bone matrix formation and calcification are linked. In other words, in the normal adult skeleton, the coupling of bone resorption and formation in remodeling results in equal levels of cellular activity so that bone turnover is balanced: the volume of bone resorbed is equal to the volume formed. This paradigm implies that, for example, a reduction in osteoblast activity would affect a similar reduction in osteoclast activity such that bone volume is maintained. Conversely, an increase in osteoclast activity should be compensated by an increase in osteoblasts and bone formation, resulting in a maintained bone mass with a high turnover, as in hyperparathyroidism for instance. Similarly, decreased osteoclast numbers or bone resorption activity should be associated with a decrease in bone formation, maintaining bone mass but with a decreased turnover rate.

 

Although this “coupling” may indeed function in most cases, there are multiple examples of dysfunctions, such as in osteoporosis or osteopetrosis for instance. It now appears that the number of osteoclasts rather than their strict activity is a key determinant of subsequent bone formation. This suggests that factors generated locally by the osteoclast, either directly or through resorption of the bone matrix, are capable of stimulating bone formation (19).

 

Haversian vs Endosteal Bone Remodeling

 

As previously mentioned, although cortical bone is anatomically different to trabecular bone, its remodeling occurs following the same sequence of events. The major difference is that while the average thickness of a trabecula is 150-200 microns, the average thickness of the cortex is of the order of 1-10 mm. There are no blood vessels in the trabeculae but the bone envelope system and the osteocyte network are able to carry out enough gaseous exchange, being always relatively close to the surface and the highly vascularized marrow. Consequently, bone remodeling in the trabecular bone will take place along the trabecular surface. On the other hand, the cortical bone itself needs to be vascularized. Blood vessels are first embedded during the histogenesis of cortical bone; the blood vessel and the bone which surrounds it is then called a primary osteon. Later, cortical bone remodeling will be initiated either along the surface of these vascular channels, or from the endosteal surface of the cortex. The remodeling process in cortical bone also follows the ARF sequence. Osteoclasts excavate a tunnel, creating a cutting cone. Again, there is a reversal phase, where mononuclear cells attach and lay down a cement line. Osteoblasts are then responsible for closing the cone, leaving a central canal, centered on blood vessels and surrounded by concentric bone lamellae. For mechanical reasons, all these Haversian systems are oriented along the longitudinal axis of the bone.

 

Bone Turnover and Skeletal Homeostasis

 

In a normal young adult, about 30% of the total skeletal mass is renewed every year (half-life = 20 months). In each remodeling unit, osteoclastic bone resorption lasts about 3 days, the reversal 14 days, and bone formation 70 days (total = 87 days). The linear bone formation rate is 0.5mm/day. During this process, about 0.01mm of bone is renewed in one given remodeling unit. Theoretically, with balanced matrix deposition and calcification as well as a balance between osteoclast and osteoblast activity, the amount of bone formed in each remodeling unit (and therefore in the total skeleton) equals the amount of bone which was previously resorbed. Thus, the total skeletal mass remains constant. This skeletal homeostasis relies upon a normal remodeling activity. The rate of activation of new remodeling units would then determine only the turnover rate.

 

SKELETAL DEVELOPMENT-HISTOGENESIS 

 

Bone development is achieved through the use of two distinct processes, intramembranous and endochondral bone formation. In the first, mesenchymal cells differentiate directly into osteoblasts whereas in the second mesenchymal cells differentiate into chondrocytes and it is only secondarily that osteoblasts appear and form bone around the cartilage model. Through a process that involves bone resorption by osteoclasts, vascular invasion and resorption of calcified cartilage, the cartilage model is progressively replaced by osteoblast-derived bone matrix. Bone is then remodeled through continuous cycles of bone resorption and formation, thereby allowing shape changes and adaptation to the local and systemic environment.

 

Intramembranous Ossification

 

During intramembranous ossification, a group of mesenchymal cells within a highly vascularized area of the embryonic connective tissue proliferates, forming early mesenchymal condensations within which cells differentiate directly into osteoblasts. Bone Morphogenetic Proteins, as well as FGFs appear to be essential in the process of mesenchymal cell condensation. The newly differentiated osteoblasts will synthesize a woven bone matrix, while at the periphery, mesenchymal cells continue to differentiate into osteoblasts. Blood vessels are incorporated between the woven bone trabeculae and will form the hematopoietic bone marrow. Later this woven bone will be remodeled through the classical remodeling process, resorbing woven bone and progressively replacing it with mature lamellar bone.

 

Endochondral Ossification

 

Development of long bones begins with the formation of a cartilage anlage (model) from a mesenchymal condensation, as in intramembranous ossification. (Figure 9). But here, under the influence of a different set of factors and local conditions, mesenchymal cells undergo division and differentiate into prechondroblasts and then into chondroblasts rather than directly into osteoblasts. These cells secrete the cartilaginous matrix, where the predominant collagen type is collagen type II. Like osteoblasts, the chondroblasts become progressively embedded within their own matrix, where they lie within lacunae, and they are then called chondrocytes. Unlike osteocytes however, chondrocytes continue to proliferate for some time, this being allowed in part by the gel-like consistency of cartilage. At the periphery of this cartilage (the perichondrium), the mesenchymal cells continue to proliferate and differentiate through appositional growth. Another type of growth is observed in the cartilage by cell proliferation and synthesis of new matrix between the chondrocytes (interstitial growth).

Figure 9. Duration and depth of the phases of the normal cancellous bone remodeling sequence, calculated from histomorphometric analysis of bone biopsy samples from young individuals (Adapted from: Eriksen EF, Axelrod DW, Melsen F. Bone Histomorphometry. Raven Press, New York, pp13-20, 1994).

 

Beginning in the center of the cartilage model, at what is to become the primary ossification center, chondrocytes continue to differentiate and become hypertrophic. During this process, hypertrophic cells deposit a mineralized matrix, where cartilage calcification is initiated by matrix vesicles. Once this matrix is calcified, it is partially resorbed by osteoclasts. After resorption and a reversal phase, osteoblasts differentiate in this area and form a layer of woven bone on top of the remaining cartilage. This woven bone will later be remodeled into lamellar bone.

 

Chondrocyte differentiation is regulated by a number of factors which have recently been described. The first factor shown to control chondrocyte differentiation was parathyroid hormone related peptide (PTHrP) acting on PTH receptors mostly found in prehypertrophic chondrocytes. This factor prolongs chondrocyte proliferation, and in PTHrP knockout mice, the main phenotype is bone shortening caused by premature chondrocyte hypertrophy. Targeted overexpression of PTHrP results in the opposite phenotype, with prolonged delay in chondrocyte maturation. PTHrP is part of a genetic signaling cascade, where not only is it regulated by factors expressed earlier in chondrocyte differentiation, such as Indian hedgehog (Ihh), but it also regulates chondrocyte differentiation itself, and alters gene expression in more mature chondrocytes. Other factors which regulate chondrocyte differentiation include the FGFs and bone morphogenetic proteins (BMPs). The transcription factors Runx2 and Sox9, together with the Wnt signaling pathway, control the commitment and differentiation within the chondrocytic lineage (20).

 

The embryonic cartilage is avascular. During its early development, a ring of woven bone is formed, the bone collar, at the periphery by intramembranous ossification in the future midshaft area under the perichondrium (which becomes periosteum). Following calcification of this woven bone, blood vessels, preceded by osteoclasts enter the primary ossification center, penetrate the bone collar and the calcified cartilage, to form the blood supply and allow seeding of the hematopoietic bone marrow. The osteoclast invasion and its concomitant wave of resorbing activity leads to the removal of the calcified cartilage and its replacement by woven bone in the primary spongiosa, as described above.

 

Secondary ossification centers begin to form at the epiphyseal ends of the cartilaginous model, and by a similar process, trabecular bone and a marrow space are formed. Between the primary and secondary ossification centers, epiphyseal cartilage (growth plates) remain until adulthood. The continued differentiation of chondrocytes, cartilage mineralization and subsequent remodeling cycles allow longitudinal bone growth to occur, such that as new bone is formed the bone will reach its final adult shape. There is, however, a progressive decrease in chondrocyte proliferation so that the growth plate becomes progressively thinner, allowing mineralization and resorption to catch up. It is at this point that the growth plates are completely remodeled and longitudinal growth is arrested.

 

The growth plate demonstrates, from the epiphyseal area to the diaphyseal area, the different stages of chondrocyte differentiation involved in endochondral bone formation (Figure 10). Firstly, a proliferative zone, where the chondroblasts divide actively, forming isogenous groups, and actively synthesizing the matrix. These cells become progressively larger, enlarging their lacunae in the pre-hypertrophic and hypertrophic zones. Lower in this area, the matrix of the longitudinal cartilage septa selectively calcifies (zone of provisional calcification). The chondrocytes become highly vacuolated and then die through programmed cell death (apoptosis). Once calcified, the cartilage matrix is resorbed, but only partially, by osteoclasts, leaving the calcified longitudinal septae and blood vessels appear in the zone of invasion. After resorption, osteoblasts differentiate and form a layer of woven bone on top of the cartilaginous remnants of the longitudinal septa. Thus, the first remodeling sequence is complete: the cartilage has been remodeled and replaced by woven bone. The resulting trabeculae are called the primary spongiosa. Still lower in the growth plate, this woven bone is subjected to further remodeling (a second ARF sequence) in which the woven bone and the cartilaginous remnants are replaced with lamellar bone, resulting in the mature trabecular bone called secondary spongiosum.

Figure 10. Bone Development. Schematic diagram showing the initial stages of endochondral ossification. Bone development begins with mesenchymal condensation to form a cartilage model of the bone to be formed. Following chondrocyte hypertrophy and cartilage matrix mineralization, osteoclast activity and vascularization result in the formation of the primary, and then secondary ossification centers. In mature adult bones, the growth plate is fully resorbed, so that one marrow cavity extends the full length of the bone. See text for details.

 

GROWTH IN BONE SHAPE AND DIAMETER (MODELING)

 

During longitudinal growth, and due to the fact that the midshaft is narrower than the metaphysis, the growth of a long bone progressively destroys the lower part of the metaphysis and transforms it into a diaphysis, a process accomplished by continuous resorption by osteoclasts beneath the periosteum.

In contrast, growth in the diameter of the metaphysis is the result of a deposition of new membranous bone beneath the periosteum that will continue throughout life. In this case, resorption does not immediately precede formation. Recently, more attention has been focusing on this type of bone formation inasmuch as periosteal bone formation seems to respond differently and/or independently from endosteal bone formation activity to different stimuli such as PTH or biomechanical loading. This is particularly important in the context of osteoporosis where it has been demonstrated that growth in diameter in the midshaft is a more important contributor to the decrease in the fracture risk than trabecular bone density and/or cortical thickness.

 

REFERENCES

 

  1. Fukumoto S, Martin TJ. Bone as an endocrine organ. Trends Endocrinol Metab. 2009 Jul;20(5):230-6.
  2. Zanatta LC, Boguszewski CL, Borba VZ, Kulak CA. Osteocalcin, energy and glucose metabolism. Arq Bras Endocrinol Metabol. 2014 Jul;58(5):444-51
  3. Ahmed LA, Shigdel R, Joakimsen RM, Eldevik OP, Eriksen EF, Ghasem-Zadeh A, Bala Y, Zebaze R, Seeman E, Bjørnerem Å. Measurement of cortical porosity of the proximal femur improves identification of women with nonvertebral fragility fractures. Osteoporos Int. 2015 Aug;26(8):2137-46.
  4. Robey P.G., Bone Matrix Proteoglycans and Glycoproteins, In Principles of Bone Biology, J.P. Bilezikian, L.G. Raisz and G.A. rodan Editors, Academic Press Publishers, Chapter 14, 225-238, 2002.
  5. Bonewald L.F., Transforming Growth Factor-β, In Principles of Bone Biology, J.P. Bilezikian, L.G. Raisz and G.A. rodan Editors, Academic Press Publishers, Chapter 49, 903-918, 2002.
  6. Harada S. and Rodan G.A., Control of osteoblast function and regulation of bone mass, Nature 423, 349-355, 2003.
  7. Baron R. and Rawadi G., Targeting the Wnt/β-catenin Pathway to Regulate Bone Formation in the Adult Skeleton, Endocrinology 148, 2635-43.
  8. Boyle W.J., Simonet W.S., Lacey D.L., Osteoclast differentiation and activation, Nature 423, 337 – 342, 2003.
  9. Asagiri M. and Takayanagi H., The molecular understanding of osteoclast differentiation, Bone, 40:251-264, 2007.
  10. Teitelbaum, S.L. & Ross, F.P., Genetic regulation of osteoclast development and function, Nature Reviews Genetics, 4, 638-649, 2003
  11. Bruzzaniti A. and Baron R. Molecular regulation of osteoclast activity. Rev. Endocr. Metab. Disord. 2007, 7:123-39.
  12. Destaing O., Saltel F., Geminard J.C., Jurdic P. and Bard F. Podosomes display actin turn-over and dynamic self-organization in osteoclasts expressing actin-GFP. Mol. Biol. Cell, 14, 407-416, 2003.
  13. Courtial N, Smink JJ, Kuvardina ON, Leutz A, Göthert JR, Lausen J. Tal1 regulates osteoclast differentiation through suppression of the master regulator of cell fusion DC-STAMP. FASEB J. 2012 Feb;26(2):523-32.
  14. Islam R, Bae HS, Yoon WJ, Woo KM, Baek JH, Kim HH, Uchida T, Ryoo HM. Pin1 regulates osteoclast fusion through suppression of the master regulator of cell fusion DC-STAMP. J Cell Physiol. 2014 Dec;229(12):2166-74.
  15. Islam R, Yoon WJ, Ryoo HM. Pin1, the Master Orchestrator of Bone Cell Differentiation. J Cell Physiol. 2017 Sep;232(9):2339-2347.
  16. Takayanagi H., Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems, Nature Reviews Immunology 7, 292 – 304, 2007.
  17. Dempster DW. Tethering Formation to Resorption: Reversal Revisited. J Bone Miner Res. 2017 Jul;32(7):1389-1390.
  18. Lassen NE, Andersen TL, Pløen GG, Søe K, Hauge EM, Harving S, Eschen GET, Delaisse JM. Coupling of Bone Resorption and Formation in Real Time: New Knowledge Gained From Human Haversian BMUs. J Bone Miner Res. 2017 Jul;32(7):1395-1405.
  19. Sims NA, Martin TJ. Coupling Signals between the Osteoclast and Osteoblast: How are Messages Transmitted between These Temporary Visitors to the Bone Surface? Front Endocrinol (Lausanne). 2015 Mar 24; 6:41.

20.. Kronenberg H.M., Developmental regulation of the growth plate, Nature 332-336, 2003.

 

 

Sitosterolemia

ABSTRACT

Sitosterolemia is a rare autosomal recessive disorder of non-cholesterol sterol metabolism, caused by mutations of the ABCG5 or ABCG8 transporter genes. This results in hyperabsorption and decreased biliary excretion of non-cholesterol sterol, especially sitosterol, from the gastrointestinal tract.  Affected individuals have excessive accumulation of plant sterols and 5 alpha-saturated stanols in plasma and tissues, resulting in premature cardiovascular disease. The condition is often clinically confused with familial hypercholesterolemia. This article provided overview of this rare condition, including diagnostic evaluation and treatment.

 

BACKGROUND

Sterols are waxy insoluble substances and are synthesized from acetyl coenzyme A (CoA).  Perhaps the most familiar example is cholesterol. In addition to cholesterol, over forty non-cholesterol sterols are also present in the human diet. Non-cholesterol sterols are contained in plants, fungi, and yeast. Instead of converting squalene to cholesterol, non-cholesterol sterols occur when squalene is converted to stigmasterol, sitosterol, campesterol, ergosterol, etc., while shellfish produce fucosterol. 

In a typical Western diet, plant sterols, or phytosterols, are often consumed in nuts, seeds, legumes, and vegetable oils. They are present in amounts equal to cholesterol and processed by the intestine in a similar manner (Figure 1).  While most individuals absorb, on average, 40-50% of dietary cholesterol, less than 5% of dietary plant sterols are absorbed (1-3).

Figure 1. Enterocyte Trafficking of Cholesterol and Plant Sterols. From Phytoserolemia by Thomas Daysring, MD in Therapeutic Lipidology, Michael H Davis in, MD, Peter P Toth, MD and Kevin C Maki, PhD, Editors. 2007 Humana Press, Incorp. Totowa, New Jersey.

Phytosterols have no role in human metabolism.  Therefore, except in inherited disorders of metabolism, there is limited systemic absorption of phytosterols, as their entry into the plasma is highly regulated by the intestine and liver. Concentrations of phystosterols in plasma are normally less than 0.5% that of cholesterol. 

Stanols, i.e., saturated sterols, also exist in the diet, primarily from plant sources.  Stanols are not normally absorbed from the GI tract. Both stanols and sterols interfere with the absorption of cholesterol. Therefore, both have been used as dietary supplements for over 5 decades to help reduce plasma cholesterol levels.

Phytosterols and free cholesterol are normally absorbed by the Niemann-Pick C1-Like 1 (NPC1L1) protein expressed on enterocytes (Figure 1) (4).  Almost all of the absorbed plant phytosterols are excreted back into the intestinal lumen by the ABCG5 or ABCG8 transporters.   The normal body is thus able to discriminate between cholesterol and non-cholesterol sterols (5). The function of ABCG5 or ABCG8 transporter genes, found at the STSL locus of human chromosome 2p21, is to limit intestinal absorption and promote biliary excretion (6, 7) (Figure 2).   

Figure 2. Normal Intestinal and Hepatic Transport of Cholesterol and Phytosterols. T. Plösch, A. Kosters, A.K. Groen, F. Kuipers. The ABC of Hepatic and Intestinal Cholesterol Transport. Chapter. Atherosclerosis: Diet and Drugs. Volume 170 of the series Handbook of Experimental Pharmacology pp 465-482.

SITOSTEROLEMIA

Sitosterolemia (also known as phytosterolemia) is a rare autosomal recessive disease of non-cholesterol sterol metabolism.  It is characterized chemically by the accumulation of plant sterols and 5 alpha-saturated stanols in plasma and tissues. The condition occurs when either ABCG5 or ABCG8 are defective, leading to hyperabsorption of sitosterol from the gastrointestinal tract.  The problem is compounded by decreased biliary excretion, resulting in accumulation of dietary phytosterols in different tissues (8, 9).

HISTORY AND ETHNICITY

Sitosterolemia was first reported in 1974 when two sisters with extensive tendon xanthomas were found to have normal plasma cholesterol levels and elevated levels of plant sterols (10). Several hundred cases have since been reported but the condition is thought to be substantially underdiagnosed (11).   The disorder has been found in a wide range of diverse populations, including the Old-Order Amish, Chinese, Finnish, Japanese, Norwegian, Indian and Caucasian South Africans, as well as others.  The condition is transmitted as an autosomal recessive trait (12, 13)

CLINICAL FEATURES

 

Signs and Symptoms

Phenotypically, sitosterolemia is very heterogeneous in its presentation. The disorder is characterized by premature coronary artery disease (14-18) although the degree of atherosclerosis present varies significantly (19-24).  Presenting signs and symptoms of sitosterolemia, such as lipid deposition in cutaneous and subcutaneous structures (xanthomas), can occur in the first decade of life, but sitosterolemia has been diagnosed in asymptomatic adults as well. Typical xanthomas occur most prominently in the extensor tendons of the hands and Achilles tendon, but can occur in the knees, elbows and buttocks. Xanthomas have been reported in children as young as one to two years of age (25-31). Spinal xanthomas, causing spinal cord compression, have also been reported (32)

The phenotype of sitosterolemia includes abnormal liver function tests, arthralgia, splenomegaly, and hematologic findings (hemolytic anemia, abnormally shaped erythrocytes and large platelets) (33-37). Occasionally, hematologic findings appear as isolated findings (11, 38-41), and there is a case report of an infant with cholestatic jaundice who was ultimately diagnosed with sitosterolemia (42).  Aortic stenosis has also been reported (21, 43), as have arthralgias and arthritis (44, 45).

Occasionally, the diagnosis of sitosterolemia is made after an individual with total cholesterol and LDL-cholesterol in the range of familial hypercholesterolemia fails to achieve expected reductions with statin therapy (46).  A recent study of 220 hypercholesterolemic children found that 6.4% had elevated and 1.4% had markedly elevated sitosterol levels, with 2 children ultimately diagnosed with genetically confirmed sitosterolemia (47).  This has been demonstrated in other publications as well (48, 49).  This reaffirms that sitosterolemia is likely underdiagnosed, and high clinical suspicion is warranted.  This is particularly important as most genetic testing panels for familial hypercholesterolemia test for pathogenic variants in LDLR, APOB, PCSK9, and LDLRAP1; therefore, individuals with sitosterolemia will frequently have negative genetic testing results.

Although sitosterolemia is a recessive disorder, there is some data suggesting that heterozygous carriers of loss of function mutations can have higher sitosterol levels, higher LDL-cholesterol levels, and a 2-fold higher risk of ASCVD (50).

Differential Diagnosis

Besides sitosterolemia, other disorders that cause tendon xanthomas in children and adults include:

Heterozygous familial hypercholesterolemia (HeFH) - most commonly caused by a co-dominantly inherited disorder of the LDL-C receptor, presents with high total serum and LDL-cholesterol, normal plasma levels of plant sterols and at least one parent with hypercholesterolemia.

Homozygous familial hypercholesterolemia (HoFH) - in which hypercholesterolemia is present in both parents of an affected child. In addition, individuals with HoFH have normal rather than enlarged platelets (macrothrombocytopenia).

Cerebrotendinous xanthomatosis (CTX) - can be distinguished by increased concentrations of plasma cholestanol, protracted diarrhea starting in childhood, and juvenile cataracts. Adults with CTX typically have neurologic involvement (cerebellar ataxia, cognitive decline, and dementia).

 

Alagille Syndrome, is accompanied by a characteristic syndromic facial appearance, high rates of congenital heart disease, and signs of liver cholestasis (51).

 

Sitosterolemia should be considered in a child or adult with tendon xanthomas and unexplained hemolysis and/or macrothrombocytopenia, as these hematologic abnormalities are not present in FH, CTX or Alagille syndrome.

Testing

Routine laboratory methods do not always distinguish plant sterols from cholesterol. Detection of plant sterol levels in blood requires gas-liquid chromatography (GLC), gas chromatography/mass spectrometry (GC/MS), or high-pressure liquid chromatography (HPLC).

Plant sterols, especially sitosterol, and the 5-alpha derivatives of plant sterols, are dramatically elevated in patients with sitosterolemia. Plasma concentrations of sitosterol above 1 mg/dL (10µg/mL) are considered to be diagnostic, although a recent study suggested a cutoff value of 15µg/mL had higher positive predictive value (52). Levels typically range from 8-60 mg/dL, 10-25 times higher than normal individuals. Age-dependent reference intervals for phytosterols have also been proposed (53). Molecular genetic testing of mutations in ABCG5 and ABCG8 can help confirm the diagnosis and direct clinical care (54).

In contrast to the very high levels of plant sterols in adults and adolescents with sitosterolemia, total cholesterol levels are sometimes normal or only moderately elevated (34). However, at least three cases of breastfed infants with sitosterolemia presenting with very elevated serum cholesterol levels have been reported. The mechanism of exceptionally high cholesterol levels in sitosterolemic children is unclear (25, 26, 55).

Increased plasma concentrations of plant sterols (especially sitosterol, campesterol, and stigmasterol) are only observed once foods with plant sterols are included in the diet and accumulate in the body. Care must be taken when evaluating infants, since commercial formula feedings with large amounts of vegetable oil may result in elevated sitosterol levels (56).

Children with parenteral nutrition associated cholestasis may have plasma concentrations of plant sterols as high as those seen in patients with hereditary sitosterolemia (i.e., total plant phytosterols of 1.3-1.8 mmol/L). Intralipid typically contains cholesterol, sitosterol, campesterol, and stigmasterol, the latter three of which are plant sterols. Adults receiving parenteral nutrition may also have elevated plasma plant sterol levels (57).

MANAGEMENT OF SITOSTEROLEMIA

 

Dietary Treatment

Treatment includes dietary restriction of non-cholesterol sterols, limiting intake of shellfish (clams, scallops, oysters), plant foods that contain high fats, such as olives, margarine, nuts, seeds, avocados, and chocolate, and avoidance of vegetable fats and oils (10, 58-61).  Fruits, vegetables and cereal products without germ may be used, however (62).

In homozygotes, plasma sterol levels may not improve significantly despite significant dietary sitosterol restriction (63, 64). Margarines and other products containing stanols (e.g., campestanol and sitostanol), which are recommended for use by individuals with hypercholesterolemia, are contraindicated in those with sitosterolemia as they can exacerbate plant stanol accumulation (65).

 

Medical Treatments

Ezetimibe (Zetia®), inhibits NPC1L1 and decreases the absorption of sterols.  It is the first-line drug therapy, lowering plant sterols by 10 to 50% and may stabilize xanthomas (66-69). Hemolytic anemia and platelet abnormalities have been reported to improve as well (66).

Bile acid sequestrants, such as cholestyramine (8-15 g/d), may be considered in those with an incomplete response to ezetimibe(26)  Regression of xanthomas has been reported in an 11-year-old after treatment with diet and cholestyramine (70). A 60-year-old man with compound heterozygous mutations in ABCG5 responded to a combination of ezetimibe and alirocumab (71).

Sitosterolemic patients do not have expected clinical responses to statins, which can help to distinguish these patients with elevated plasma sterols and xanthomas from those with familial hypercholesterolemia (64).  As stated above, sitosterolemia should be suspected in individuals with hypercholesterolemia who fail to respond as expected to a statin treatment.

 

Surgical Treatments

 

Partial ileal bypass surgery (i.e., shortening of the ileum) has been used to increase intestinal bile acid loss. Partial or complete ileal bypass surgery in persons with sitosterolemia has resulted in at least 50% reduction of plasma and cellular sterol and stanol levels (72-74).

Surgical treatments for complications of sitosterolemia have been reported. Liver cirrhosis has been observed at least once in a patient with the ABCG8 mutation. The patient underwent successful treatment by liver transplant, which led to a dramatic improvement in the sitosterolemia. It is possible that restoration of the ABCG8 function in the liver alone may be sufficient to correct the biochemical abnormality (22).

REFERENCES

  1. Gould RG, Jones RJ, LeRoy GV, Wissler RW, Taylor CB. Absorbability of beta-sitosterol in humans. Metabolism. 1969;18(8):652-62.
  2. Salen G, Ahrens EH, Jr., Grundy SM. Metabolism of beta-sitosterol in man. J Clin Invest. 1970;49(5):952-67.
  3. Salen G, Tint GS, Shefer S, Shore V, Nguyen L. Increased sitosterol absorption is offset by rapid elimination to prevent accumulation in heterozygotes with sitosterolemia. Arterioscler Thromb. 1992;12(5):563-8.
  4. Davis HR, Jr., Zhu LJ, Hoos LM, Tetzloff G, Maguire M, Liu J, et al. Niemann-Pick C1 Like 1 (NPC1L1) is the intestinal phytosterol and cholesterol transporter and a key modulator of whole-body cholesterol homeostasis. J Biol Chem. 2004;279(32):33586-92.
  5. Patel SB, Honda A, Salen G. Sitosterolemia: exclusion of genes involved in reduced cholesterol biosynthesis. J Lipid Res. 1998;39(5):1055-61.
  6. Patel SB, Salen G, Hidaka H, Kwiterovich PO, Stalenhoef AF, Miettinen TA, et al. Mapping a gene involved in regulating dietary cholesterol absorption. The sitosterolemia locus is found at chromosome 2p21. The Journal of clinical investigation. 1998;102(5):1041-4.
  7. Patel SB. Plant sterols and stanols: their role in health and disease. J Clin Lipidol. 2008;2(2):S11-S9.
  8. Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, et al. Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science. 2000;290(5497):1771-5.
  9. Wang J, Mitsche MA, Lutjohann D, Cohen JC, Xie XS, Hobbs HH. Relative roles of ABCG5/ABCG8 in liver and intestine. J Lipid Res. 2015;56(2):319-30.
  10. Bhattacharyya AK, Connor WE. β-Sitosterolemia and Xanthomatosis: A NEWLY DESCRIBED LIPID STORAGE DISEASE IN TWO SISTERS. The Journal of Clinical Investigation. 1974;53(4):1033-43.
  11. Escola-Gil JC, Quesada H, Julve J, Martin-Campos JM, Cedo L, Blanco-Vaca F. Sitosterolemia: diagnosis, investigation, and management. Current atherosclerosis reports. 2014;16(7):424.
  12. Beaty TH, Kwiterovich PO, Jr., Khoury MJ, White S, Bachorik PS, Smith HH, et al. Genetic analysis of plasma sitosterol, apoprotein B, and lipoproteins in a large Amish pedigree with sitosterolemia. Am J Hum Genet. 1986;38(4):492-504.
  13. Lee MH, Lu K, Patel SB. Genetic basis of sitosterolemia. Curr Opin Lipidol. 2001;12(2):141-9.
  14. Kidambi S, Patel SB. Sitosterolaemia: pathophysiology, clinical presentation and laboratory diagnosis. J Clin Pathol. 2008;61(5):588-94.
  15. Patil S, Kharge J, Bagi V, Ramalingam R. Tendon xanthomas as indicators of atherosclerotic burden on coronary arteries. Indian Heart J. 2013;65(4):491-2.
  16. Salen G, Horak I, Rothkopf M, Cohen JL, Speck J, Tint GS, et al. Lethal atherosclerosis associated with abnormal plasma and tissue sterol composition in sitosterolemia with xanthomatosis. J Lipid Res. 1985;26(9):1126-33.
  17. Bhattacharyya AK, Connor WE, Lin DS, McMurry MM, Shulman RS. Sluggish sitosterol turnover and hepatic failure to excrete sitosterol into bile cause expansion of body pool of sitosterol in patients with sitosterolemia and xanthomatosis. Arterioscler Thromb. 1991;11(5):1287-94.
  18. Kolovou G, Voudris V, Drogari E, Palatianos G, Cokkinos DV. Coronary bypass grafts in a young girl with sitosterolemia. Eur Heart J. 1996;17(6):965-6.
  19. Hansel B, Carrie A, Brun-Druc N, Leclert G, Chantepie S, Coiffard AS, et al. Premature atherosclerosis is not systematic in phytosterolemic patients: severe hypercholesterolemia as a confounding factor in five subjects. Atherosclerosis. 2014;234(1):162-8.
  20. Mymin D, Salen G, Triggs-Raine B, Waggoner DJ, Dembinski T, Hatch GM. The natural history of phytosterolemia: Observations on its homeostasis. Atherosclerosis. 2018;269:122-8.
  21. Wang J, Joy T, Mymin D, Frohlich J, Hegele RA. Phenotypic heterogeneity of sitosterolemia. J Lipid Res. 2004;45(12):2361-7.
  22. Miettinen TA, Klett EL, Gylling H, Isoniemi H, Patel SB. Liver transplantation in a patient with sitosterolemia and cirrhosis. Gastroenterology. 2006;130(2):542-7.
  23. Yamamoto T, Matsuda J, Dateki S, Ouchi K, Fujimoto W. Numerous intertriginous xanthomas in infant: A diagnostic clue for sitosterolemia. The Journal of Dermatology. 2016;43(11):1340-4.
  24. Peterson AL, DeLine J, Korcarz CE, Dodge AM, Stein JH. Phenotypic Variability in Atherosclerosis Burden in an Old-Order Amish Family With Homozygous Sitosterolemia. JACC: Case Reports. 2020;2(4):646-50.
  25. Park JH, Chung IH, Kim DH, Choi MH, Garg A, Yoo EG. Sitosterolemia presenting with severe hypercholesterolemia and intertriginous xanthomas in a breastfed infant: case report and brief review. J Clin Endocrinol Metab. 2014;99(5):1512-8.
  26. Niu DM, Chong KW, Hsu JH, Wu TJ, Yu HC, Huang CH, et al. Clinical observations, molecular genetic analysis, and treatment of sitosterolemia in infants and children. Journal of inherited metabolic disease. 2010;33(4):437-43.
  27. Shulman RS, Bhattacharyya AK, Connor WE, Fredrickson DS. β-Sitosterolemia and Xanthomatosis. New England Journal of Medicine. 1976;294(9):482-3.
  28. Davidson CS. The Metabolic Basis of Inherited Disease. Fifth Edition. Edited by John B. Stanbury, James B. Wyn-gaarden, Donald S. Fredrickson, Joseph L. Goldstein, and Michael S. Brown, xvi + 2,032 pp. New York: McGraw-Hill, 1983. Hepatology. 1983;3(3):461-2.
  29. Kalter H. The metabolic basis of inherited disease. 3rd Ed. J. B. Stanbury, J. B. Wyngaarden, and D. S. Fredrickson, eds. McGraw-Hill, New York. 1778 pp. 1972. Teratology. 1972;6(3):362-.
  30. Hubacek JA, Berge KE, Cohen JC, Hobbs HH. Mutations in ATP-cassette binding proteins G5 (ABCG5) and G8 (ABCG8) causing sitosterolemia. Hum Mutat. 2001;18(4):359-60.
  31. Watts GF, Mitchell WD. Clinical and metabolic findings in a patient with phytosterolaemia. Ann Clin Biochem. 1992;29 ( Pt 2):231-6.
  32. Hidaka H, Yasuda H, Kobayashi M, Hatanaka I, Takahashi M, Matsumoto K, et al. Familial spinal xanthomatosis with sitosterolemia. Intern Med. 1992;31(8):1038-42.
  33. Salen G, Shefer S, Nguyen L, Ness GC, Tint GS, Shore V. Sitosterolemia. J Lipid Res. 1992;33(7):945-55.
  34. Björkhem I, Boberg KM, Leitersdorf E. Inborn Errors in Bile Acid Biosynthesis and Storage of Sterols Other than Cholesterol. In: Valle D, Antonarakis S, Ballabio A, Beaudet A, Mitchell GA, editors. The Online Metabolic and Molecular Bases of Inherited Disease. New York, NY: McGraw-Hill Education; 2019.
  35. Salen G, Patel S, Batta AK. Sitosterolemia. Cardiovasc Drug Rev. 2002;20(4):255-70.
  36. Su Y, Wang Z, Yang H, Cao L, Liu F, Bai X, et al. Clinical and molecular genetic analysis of a family with sitosterolemia and co-existing erythrocyte and platelet abnormalities. Haematologica. 2006;91(10):1392-5.
  37. Wang G, Wang Z, Liang J, Cao L, Bai X, Ruan C. A phytosterolemia patient presenting exclusively with macrothrombocytopenia and stomatocytic hemolysis. Acta Haematol. 2011;126(2):95-8.
  38. Kaya Z, Niu DM, Yorulmaz A, Tekin A, Gursel T. A novel mutation of ABCG5 gene in a Turkish boy with phytosterolemia presenting with macrotrombocytopenia and stomatocytosis. Pediatr Blood Cancer. 2014;61(8):1457-9.
  39. Wang Z, Cao L, Su Y, Wang G, Wang R, Yu Z, et al. Specific macrothrombocytopenia/hemolytic anemia associated with sitosterolemia. Am J Hematol. 2014;89(3):320-4.
  40. Gülen H, Yıldırım AT, Yiğit Y, Yorulmaz A. Typical hematological findings facilitating the diagnosis of sitosterolemia. Pediatr Int. 2021;63(4):472-3.
  41. Gok V, Tada H, Ensar Dogan M, Alakus Sari U, Aslan K, Ozcan A, et al. A teenager boy with a novel variant of Sitosterolemia presented with pancytopenia. Clin Chim Acta. 2022;529:61-6.
  42. Mandato C, Siano MA, Nazzaro L, Gelzo M, Francalanci P, Rizzo F, et al. A ZFYVE19 gene mutation associated with neonatal cholestasis and cilia dysfunction: case report with a novel pathogenic variant. Orphanet J Rare Dis. 2021;16(1):179.
  43. Wang Y, Guo YL, Dong QT, Li JJ. Severe aortic valve stenosis in a 14-year-old boy with sitosterolemia. J Clin Lipidol. 2018.
  44. Xia Y, Duan Y, Zheng W, Liang L, Zhang H, Luo X, et al. Clinical, genetic profile and therapy evaluation of 55 children and 5 adults with sitosterolemia. J Clin Lipidol. 2022;16(1):40-51.
  45. Tada H, Kojima N, Yamagami K, Takamura M, Kawashiri MA. Clinical and genetic features of sitosterolemia in Japan. Clin Chim Acta. 2022;530:39-44.
  46. Kiss S, Lee JY, Pitt J, MacGregor D, Wallace J, Marty M, et al. Dig deeper when it does not make sense: Juvenile xanthomas due to sitosterolemia. JIMD Rep. 2020;56(1):34-9.
  47. Lee JH, Song DY, Jun SH, Song SH, Shin CH, Ki CS, et al. High prevalence of increased sitosterol levels in hypercholesterolemic children suggest underestimation of sitosterolemia incidence. PLoS One. 2020;15(8):e0238079.
  48. Tada H, Okada H, Nomura A, Yashiro S, Nohara A, Ishigaki Y, et al. Rare and Deleterious Mutations in ABCG5/ABCG8 Genes Contribute to Mimicking and Worsening of Familial Hypercholesterolemia Phenotype. Circ J. 2019;83(9):1917-24.
  49. Tada MT, Rocha VZ, Lima IR, Oliveira TGM, Chacra AP, Miname MH, et al. Screening of ABCG5 and ABCG8 Genes for Sitosterolemia in a Familial Hypercholesterolemia Cascade Screening Program. Circ Genom Precis Med. 2022;15(3):e003390.
  50. Nomura A, Emdin CA, Won HH, Peloso GM, Natarajan P, Ardissino D, et al. Heterozygous ATP-binding Cassette Transporter G5 Gene Deficiency and Risk of Coronary Artery Disease. Circulation: Genomic and Precision Medicine.2020;13(5):417-423..
  51. Alagille D, Odievre M, Gautier M, Dommergues JP. Hepatic ductular hypoplasia associated with characteristic facies, vertebral malformations, retarded physical, mental, and sexual development, and cardiac murmur. J Pediatr. 1975;86(1):63-71.
  52. Kojima N, Tada H, Usui S, Sakata K, Hayashi K, Nohara A, et al. Serum sitosterol level predicting ABCG5 or ABCG8 genetic mutations. Clin Chim Acta. 2020;507:11-6.
  53. Wu M, Pei Z, Sun W, Wu H, Sun Y, Wu B, et al. Age-related reference intervals for serum phytosterols in children by gas chromatography-mass spectrometry and its application in diagnosing sitosterolemia. Clin Chim Acta. 2023;540:117234.
  54. Brown EE, Sturm AC, Cuchel M, Braun LT, Duell PB, Underberg JA, et al. Genetic testing in dyslipidemia: A scientific statement from the National Lipid Association. J Clin Lipidol. 2020;14(4):398-413.
  55. Rios J, Stein E, Shendure J, Hobbs HH, Cohen JC. Identification by whole-genome resequencing of gene defect responsible for severe hypercholesterolemia. Hum Mol Genet. 2010;19(22):4313-8.
  56. Hamdan IJA, Sanchez-Siles LM, Garcia-Llatas G, Lagarda MJ. Sterols in Infant Formulas: A Bioaccessibility Study. Journal of Agricultural and Food Chemistry. 2018;66(6):1377-85.
  57. Llop JM, Virgili N, Moreno-Villares JM, Garcia-Peris P, Serrano T, Forga M, et al. Phytosterolemia in parenteral nutrition patients: implications for liver disease development. Nutrition. 2008;24(11-12):1145-52.
  58. Kratz M, Kannenberg F, Gramenz E, Berning B, Trautwein E, Assmann G, et al. Similar serum plant sterol responses of human subjects heterozygous for a mutation causing sitosterolemia and controls to diets enriched in plant sterols or stanols. Eur J Clin Nutr. 2007;61(7):896-905.
  59. Myrie SB, Mymin D, Triggs-Raine B, Jones PJ. Serum lipids, plant sterols, and cholesterol kinetic responses to plant sterol supplementation in phytosterolemia heterozygotes and control individuals. Am J Clin Nutr. 2012;95(4):837-44.
  60. Gregg RE, Connor WE, Lin DS, Brewer HB, Jr. Abnormal metabolism of shellfish sterols in a patient with sitosterolemia and xanthomatosis. The Journal of Clinical Investigation. 1986;77(6):1864-72.
  61. Parsons HG, Jamal R, Baylis B, Dias VC, Roncari D. A marked and sustained reduction in LDL sterols by diet and cholestyramine in beta-sitosterolemia. Clin Invest Med. 1995;18(5):389-400.
  62. Tsubakio-Yamamoto K, Nishida M, Nakagawa-Toyama Y, Masuda D, Ohama T, Yamashita S. Current Therapy for Patients with Sitosterolemia --Effect of Ezetimibe on Plant Sterol Metabolism. Journal of atherosclerosis and thrombosis. 2010;17(9):891-900..
  63. Cobb MM, Salen G, Tint GS. Comparative effect of dietary sitosterol on plasma sterols and cholesterol and bile acid synthesis in a sitosterolemic homozygote and heterozygote subject. J Am Coll Nutr. 1997;16(6):605-13.
  64. Nguyen LB, Cobb M, Shefer S, Salen G, Ness GC, Tint GS. Regulation of cholesterol biosynthesis in sitosterolemia: effects of lovastatin, cholestyramine, and dietary sterol restriction. J Lipid Res. 1991;32(12):1941-8.
  65. Connor WE, Lin DS, Pappu AS, Frohlich J, Gerhard G. Dietary sitostanol and campestanol: accumulation in the blood of humans with sitosterolemia and xanthomatosis and in rat tissues. Lipids. 2005;40(9):919-23.
  66. Othman RA, Myrie SB, Mymin D, Merkens LS, Roullet JB, Steiner RD, et al. Ezetimibe reduces plant sterol accumulation and favorably increases platelet count in sitosterolemia. J Pediatr. 2015;166(1):125-31.
  67. Salen G, von Bergmann K, Lutjohann D, Kwiterovich P, Kane J, Patel SB, et al. Ezetimibe effectively reduces plasma plant sterols in patients with sitosterolemia. Circulation. 2004;109(8):966-71.
  68. Lutjohann D, von Bergmann K, Sirah W, Macdonell G, Johnson-Levonas AO, Shah A, et al. Long-term efficacy and safety of ezetimibe 10 mg in patients with homozygous sitosterolemia: a 2-year, open-label extension study. Int J Clin Pract. 2008;62(10):1499-510.
  69. Salen G, Starc T, Sisk CM, Patel SB. Intestinal cholesterol absorption inhibitor ezetimibe added to cholestyramine for sitosterolemia and xanthomatosis. Gastroenterology. 2006;130(6):1853-7.
  70. Belamarich PF, Deckelbaum RJ, Starc TJ, Dobrin BE, Tint GS, Salen G. Response to diet and cholestyramine in a patient with sitosterolemia. Pediatrics. 1990;86(6):977-81.
  71. Tanaka H, Watanabe Y, Hirano S, Tada H, Nomura A, Kawashiri MA, et al. Sitosterolemia Exhibiting Severe Hypercholesterolemia with Tendon Xanthomas Due to Compound Heterozygous ABCG5 Gene Mutations Treated with Ezetimibe and Alirocumab. Intern Med. 2020;59(23):3033-7.
  72. Nguyen LB, Shefer S, Salen G, Horak I, Tint GS, McNamara DJ. The effect of abnormal plasma and cellular sterol content and composition on low density lipoprotein uptake and degradation by monocytes and lymphocytes in sitosterolemia with xanthomatosis. Metabolism. 1988;37(4):346-51.
  73. Nguyen LB, Shefer S, Salen G, Ness GC, Tint GS, Zaki FG, et al. A molecular defect in hepatic cholesterol biosynthesis in sitosterolemia with xanthomatosis. The Journal of Clinical Investigation. 1990;86(3):923-31.
  74. Salen G, Batta AK, Tint GS, Shefer S, Ness GC. Inverse relationship between plasma cholestanol concentrations and bile acid synthesis in sitosterolemia. J Lipid Res. 1994;35(10):1878-87.

Sitosterolemia

ABSTRACT

Sitosterolemia is a rare autosomal recessive disorder of non-cholesterol sterol metabolism, caused by mutations of the ABCG5 or ABCG8 transporter genes. This results in hyperabsorption and decreased biliary excretion of non-cholesterol sterol, especially sitosterol, from the gastrointestinal tract.  Affected individuals have excessive accumulation of plant sterols and 5 alpha-saturated stanols in plasma and tissues, resulting in premature cardiovascular disease. The condition is often clinically confused with familial hypercholesterolemia. This article provided overview of this rare condition, including diagnostic evaluation and treatment.

 

BACKGROUND

Sterols are waxy insoluble substances and are synthesized from acetyl coenzyme A (CoA).  Perhaps the most familiar example is cholesterol. In addition to cholesterol, over forty non-cholesterol sterols are also present in the human diet. Non-cholesterol sterols are contained in plants, fungi, and yeast. Instead of converting squalene to cholesterol, non-cholesterol sterols occur when squalene is converted to stigmasterol, sitosterol, campesterol, ergosterol, etc., while shellfish produce fucosterol. 

In a typical Western diet, plant sterols, or phytosterols, are often consumed in nuts, seeds, legumes, and vegetable oils. They are present in amounts equal to cholesterol and processed by the intestine in a similar manner (Figure 1).  While most individuals absorb, on average, 40-50% of dietary cholesterol, less than 5% of dietary plant sterols are absorbed (1-3).

Figure 1. Enterocyte Trafficking of Cholesterol and Plant Sterols. From Phytoserolemia by Thomas Daysring, MD in Therapeutic Lipidology, Michael H Davis in, MD, Peter P Toth, MD and Kevin C Maki, PhD, Editors. 2007 Humana Press, Incorp. Totowa, New Jersey.

Phytosterols have no role in human metabolism.  Therefore, except in inherited disorders of metabolism, there is limited systemic absorption of phytosterols, as their entry into the plasma is highly regulated by the intestine and liver. Concentrations of phystosterols in plasma are normally less than 0.5% that of cholesterol. 

Stanols, i.e., saturated sterols, also exist in the diet, primarily from plant sources.  Stanols are not normally absorbed from the GI tract. Both stanols and sterols interfere with the absorption of cholesterol. Therefore, both have been used as dietary supplements for over 5 decades to help reduce plasma cholesterol levels.

Phytosterols and free cholesterol are normally absorbed by the Niemann-Pick C1-Like 1 (NPC1L1) protein expressed on enterocytes (Figure 1) (4).  Almost all of the absorbed plant phytosterols are excreted back into the intestinal lumen by the ABCG5 or ABCG8 transporters.   The normal body is thus able to discriminate between cholesterol and non-cholesterol sterols (5). The function of ABCG5 or ABCG8 transporter genes, found at the STSL locus of human chromosome 2p21, is to limit intestinal absorption and promote biliary excretion (6, 7) (Figure 2).   

Figure 2. Normal Intestinal and Hepatic Transport of Cholesterol and Phytosterols. T. Plösch, A. Kosters, A.K. Groen, F. Kuipers. The ABC of Hepatic and Intestinal Cholesterol Transport. Chapter. Atherosclerosis: Diet and Drugs. Volume 170 of the series Handbook of Experimental Pharmacology pp 465-482.

SITOSTEROLEMIA

Sitosterolemia (also known as phytosterolemia) is a rare autosomal recessive disease of non-cholesterol sterol metabolism.  It is characterized chemically by the accumulation of plant sterols and 5 alpha-saturated stanols in plasma and tissues. The condition occurs when either ABCG5 or ABCG8 are defective, leading to hyperabsorption of sitosterol from the gastrointestinal tract.  The problem is compounded by decreased biliary excretion, resulting in accumulation of dietary phytosterols in different tissues (8, 9).

HISTORY AND ETHNICITY

Sitosterolemia was first reported in 1974 when two sisters with extensive tendon xanthomas were found to have normal plasma cholesterol levels and elevated levels of plant sterols (10). Several hundred cases have since been reported but the condition is thought to be substantially underdiagnosed (11).   The disorder has been found in a wide range of diverse populations, including the Old-Order Amish, Chinese, Finnish, Japanese, Norwegian, Indian and Caucasian South Africans, as well as others.  The condition is transmitted as an autosomal recessive trait (12, 13)

CLINICAL FEATURES

 

Signs and Symptoms

Phenotypically, sitosterolemia is very heterogeneous in its presentation. The disorder is characterized by premature coronary artery disease (14-18) although the degree of atherosclerosis present varies significantly (19-24).  Presenting signs and symptoms of sitosterolemia, such as lipid deposition in cutaneous and subcutaneous structures (xanthomas), can occur in the first decade of life, but sitosterolemia has been diagnosed in asymptomatic adults as well. Typical xanthomas occur most prominently in the extensor tendons of the hands and Achilles tendon, but can occur in the knees, elbows and buttocks. Xanthomas have been reported in children as young as one to two years of age (25-31). Spinal xanthomas, causing spinal cord compression, have also been reported (32)

The phenotype of sitosterolemia includes abnormal liver function tests, arthralgia, splenomegaly, and hematologic findings (hemolytic anemia, abnormally shaped erythrocytes and large platelets) (33-37). Occasionally, hematologic findings appear as isolated findings (11, 38-41), and there is a case report of an infant with cholestatic jaundice who was ultimately diagnosed with sitosterolemia (42).  Aortic stenosis has also been reported (21, 43), as have arthralgias and arthritis (44, 45).

Occasionally, the diagnosis of sitosterolemia is made after an individual with total cholesterol and LDL-cholesterol in the range of familial hypercholesterolemia fails to achieve expected reductions with statin therapy (46).  A recent study of 220 hypercholesterolemic children found that 6.4% had elevated and 1.4% had markedly elevated sitosterol levels, with 2 children ultimately diagnosed with genetically confirmed sitosterolemia (47).  This has been demonstrated in other publications as well (48, 49).  This reaffirms that sitosterolemia is likely underdiagnosed, and high clinical suspicion is warranted.  This is particularly important as most genetic testing panels for familial hypercholesterolemia test for pathogenic variants in LDLR, APOB, PCSK9, and LDLRAP1; therefore, individuals with sitosterolemia will frequently have negative genetic testing results.

Although sitosterolemia is a recessive disorder, there is some data suggesting that heterozygous carriers of loss of function mutations can have higher sitosterol levels, higher LDL-cholesterol levels, and a 2-fold higher risk of ASCVD (50).

Differential Diagnosis

Besides sitosterolemia, other disorders that cause tendon xanthomas in children and adults include:

Heterozygous familial hypercholesterolemia (HeFH) - most commonly caused by a co-dominantly inherited disorder of the LDL-C receptor, presents with high total serum and LDL-cholesterol, normal plasma levels of plant sterols and at least one parent with hypercholesterolemia.

Homozygous familial hypercholesterolemia (HoFH) - in which hypercholesterolemia is present in both parents of an affected child. In addition, individuals with HoFH have normal rather than enlarged platelets (macrothrombocytopenia).

Cerebrotendinous xanthomatosis (CTX) - can be distinguished by increased concentrations of plasma cholestanol, protracted diarrhea starting in childhood, and juvenile cataracts. Adults with CTX typically have neurologic involvement (cerebellar ataxia, cognitive decline, and dementia).

 

Alagille Syndrome, is accompanied by a characteristic syndromic facial appearance, high rates of congenital heart disease, and signs of liver cholestasis (51).

 

Sitosterolemia should be considered in a child or adult with tendon xanthomas and unexplained hemolysis and/or macrothrombocytopenia, as these hematologic abnormalities are not present in FH, CTX or Alagille syndrome.

Testing

Routine laboratory methods do not always distinguish plant sterols from cholesterol. Detection of plant sterol levels in blood requires gas-liquid chromatography (GLC), gas chromatography/mass spectrometry (GC/MS), or high-pressure liquid chromatography (HPLC).

Plant sterols, especially sitosterol, and the 5-alpha derivatives of plant sterols, are dramatically elevated in patients with sitosterolemia. Plasma concentrations of sitosterol above 1 mg/dL (10µg/mL) are considered to be diagnostic, although a recent study suggested a cutoff value of 15µg/mL had higher positive predictive value (52). Levels typically range from 8-60 mg/dL, 10-25 times higher than normal individuals. Age-dependent reference intervals for phytosterols have also been proposed (53). Molecular genetic testing of mutations in ABCG5 and ABCG8 can help confirm the diagnosis and direct clinical care (54).

In contrast to the very high levels of plant sterols in adults and adolescents with sitosterolemia, total cholesterol levels are sometimes normal or only moderately elevated (34). However, at least three cases of breastfed infants with sitosterolemia presenting with very elevated serum cholesterol levels have been reported. The mechanism of exceptionally high cholesterol levels in sitosterolemic children is unclear (25, 26, 55).

Increased plasma concentrations of plant sterols (especially sitosterol, campesterol, and stigmasterol) are only observed once foods with plant sterols are included in the diet and accumulate in the body. Care must be taken when evaluating infants, since commercial formula feedings with large amounts of vegetable oil may result in elevated sitosterol levels (56).

Children with parenteral nutrition associated cholestasis may have plasma concentrations of plant sterols as high as those seen in patients with hereditary sitosterolemia (i.e., total plant phytosterols of 1.3-1.8 mmol/L). Intralipid typically contains cholesterol, sitosterol, campesterol, and stigmasterol, the latter three of which are plant sterols. Adults receiving parenteral nutrition may also have elevated plasma plant sterol levels (57).

MANAGEMENT OF SITOSTEROLEMIA

 

Dietary Treatment

Treatment includes dietary restriction of non-cholesterol sterols, limiting intake of shellfish (clams, scallops, oysters), plant foods that contain high fats, such as olives, margarine, nuts, seeds, avocados, and chocolate, and avoidance of vegetable fats and oils (10, 58-61).  Fruits, vegetables and cereal products without germ may be used, however (62).

In homozygotes, plasma sterol levels may not improve significantly despite significant dietary sitosterol restriction (63, 64). Margarines and other products containing stanols (e.g., campestanol and sitostanol), which are recommended for use by individuals with hypercholesterolemia, are contraindicated in those with sitosterolemia as they can exacerbate plant stanol accumulation (65).

 

Medical Treatments

Ezetimibe (Zetia®), inhibits NPC1L1 and decreases the absorption of sterols.  It is the first-line drug therapy, lowering plant sterols by 10 to 50% and may stabilize xanthomas (66-69). Hemolytic anemia and platelet abnormalities have been reported to improve as well (66).

Bile acid sequestrants, such as cholestyramine (8-15 g/d), may be considered in those with an incomplete response to ezetimibe(26)  Regression of xanthomas has been reported in an 11-year-old after treatment with diet and cholestyramine (70). A 60-year-old man with compound heterozygous mutations in ABCG5 responded to a combination of ezetimibe and alirocumab (71).

Sitosterolemic patients do not have expected clinical responses to statins, which can help to distinguish these patients with elevated plasma sterols and xanthomas from those with familial hypercholesterolemia (64).  As stated above, sitosterolemia should be suspected in individuals with hypercholesterolemia who fail to respond as expected to a statin treatment.

 

Surgical Treatments

 

Partial ileal bypass surgery (i.e., shortening of the ileum) has been used to increase intestinal bile acid loss. Partial or complete ileal bypass surgery in persons with sitosterolemia has resulted in at least 50% reduction of plasma and cellular sterol and stanol levels (72-74).

Surgical treatments for complications of sitosterolemia have been reported. Liver cirrhosis has been observed at least once in a patient with the ABCG8 mutation. The patient underwent successful treatment by liver transplant, which led to a dramatic improvement in the sitosterolemia. It is possible that restoration of the ABCG8 function in the liver alone may be sufficient to correct the biochemical abnormality (22).

REFERENCES

  1. Gould RG, Jones RJ, LeRoy GV, Wissler RW, Taylor CB. Absorbability of beta-sitosterol in humans. Metabolism. 1969;18(8):652-62.
  2. Salen G, Ahrens EH, Jr., Grundy SM. Metabolism of beta-sitosterol in man. J Clin Invest. 1970;49(5):952-67.
  3. Salen G, Tint GS, Shefer S, Shore V, Nguyen L. Increased sitosterol absorption is offset by rapid elimination to prevent accumulation in heterozygotes with sitosterolemia. Arterioscler Thromb. 1992;12(5):563-8.
  4. Davis HR, Jr., Zhu LJ, Hoos LM, Tetzloff G, Maguire M, Liu J, et al. Niemann-Pick C1 Like 1 (NPC1L1) is the intestinal phytosterol and cholesterol transporter and a key modulator of whole-body cholesterol homeostasis. J Biol Chem. 2004;279(32):33586-92.
  5. Patel SB, Honda A, Salen G. Sitosterolemia: exclusion of genes involved in reduced cholesterol biosynthesis. J Lipid Res. 1998;39(5):1055-61.
  6. Patel SB, Salen G, Hidaka H, Kwiterovich PO, Stalenhoef AF, Miettinen TA, et al. Mapping a gene involved in regulating dietary cholesterol absorption. The sitosterolemia locus is found at chromosome 2p21. The Journal of clinical investigation. 1998;102(5):1041-4.
  7. Patel SB. Plant sterols and stanols: their role in health and disease. J Clin Lipidol. 2008;2(2):S11-S9.
  8. Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, et al. Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science. 2000;290(5497):1771-5.
  9. Wang J, Mitsche MA, Lutjohann D, Cohen JC, Xie XS, Hobbs HH. Relative roles of ABCG5/ABCG8 in liver and intestine. J Lipid Res. 2015;56(2):319-30.
  10. Bhattacharyya AK, Connor WE. β-Sitosterolemia and Xanthomatosis: A NEWLY DESCRIBED LIPID STORAGE DISEASE IN TWO SISTERS. The Journal of Clinical Investigation. 1974;53(4):1033-43.
  11. Escola-Gil JC, Quesada H, Julve J, Martin-Campos JM, Cedo L, Blanco-Vaca F. Sitosterolemia: diagnosis, investigation, and management. Current atherosclerosis reports. 2014;16(7):424.
  12. Beaty TH, Kwiterovich PO, Jr., Khoury MJ, White S, Bachorik PS, Smith HH, et al. Genetic analysis of plasma sitosterol, apoprotein B, and lipoproteins in a large Amish pedigree with sitosterolemia. Am J Hum Genet. 1986;38(4):492-504.
  13. Lee MH, Lu K, Patel SB. Genetic basis of sitosterolemia. Curr Opin Lipidol. 2001;12(2):141-9.
  14. Kidambi S, Patel SB. Sitosterolaemia: pathophysiology, clinical presentation and laboratory diagnosis. J Clin Pathol. 2008;61(5):588-94.
  15. Patil S, Kharge J, Bagi V, Ramalingam R. Tendon xanthomas as indicators of atherosclerotic burden on coronary arteries. Indian Heart J. 2013;65(4):491-2.
  16. Salen G, Horak I, Rothkopf M, Cohen JL, Speck J, Tint GS, et al. Lethal atherosclerosis associated with abnormal plasma and tissue sterol composition in sitosterolemia with xanthomatosis. J Lipid Res. 1985;26(9):1126-33.
  17. Bhattacharyya AK, Connor WE, Lin DS, McMurry MM, Shulman RS. Sluggish sitosterol turnover and hepatic failure to excrete sitosterol into bile cause expansion of body pool of sitosterol in patients with sitosterolemia and xanthomatosis. Arterioscler Thromb. 1991;11(5):1287-94.
  18. Kolovou G, Voudris V, Drogari E, Palatianos G, Cokkinos DV. Coronary bypass grafts in a young girl with sitosterolemia. Eur Heart J. 1996;17(6):965-6.
  19. Hansel B, Carrie A, Brun-Druc N, Leclert G, Chantepie S, Coiffard AS, et al. Premature atherosclerosis is not systematic in phytosterolemic patients: severe hypercholesterolemia as a confounding factor in five subjects. Atherosclerosis. 2014;234(1):162-8.
  20. Mymin D, Salen G, Triggs-Raine B, Waggoner DJ, Dembinski T, Hatch GM. The natural history of phytosterolemia: Observations on its homeostasis. Atherosclerosis. 2018;269:122-8.
  21. Wang J, Joy T, Mymin D, Frohlich J, Hegele RA. Phenotypic heterogeneity of sitosterolemia. J Lipid Res. 2004;45(12):2361-7.
  22. Miettinen TA, Klett EL, Gylling H, Isoniemi H, Patel SB. Liver transplantation in a patient with sitosterolemia and cirrhosis. Gastroenterology. 2006;130(2):542-7.
  23. Yamamoto T, Matsuda J, Dateki S, Ouchi K, Fujimoto W. Numerous intertriginous xanthomas in infant: A diagnostic clue for sitosterolemia. The Journal of Dermatology. 2016;43(11):1340-4.
  24. Peterson AL, DeLine J, Korcarz CE, Dodge AM, Stein JH. Phenotypic Variability in Atherosclerosis Burden in an Old-Order Amish Family With Homozygous Sitosterolemia. JACC: Case Reports. 2020;2(4):646-50.
  25. Park JH, Chung IH, Kim DH, Choi MH, Garg A, Yoo EG. Sitosterolemia presenting with severe hypercholesterolemia and intertriginous xanthomas in a breastfed infant: case report and brief review. J Clin Endocrinol Metab. 2014;99(5):1512-8.
  26. Niu DM, Chong KW, Hsu JH, Wu TJ, Yu HC, Huang CH, et al. Clinical observations, molecular genetic analysis, and treatment of sitosterolemia in infants and children. Journal of inherited metabolic disease. 2010;33(4):437-43.
  27. Shulman RS, Bhattacharyya AK, Connor WE, Fredrickson DS. β-Sitosterolemia and Xanthomatosis. New England Journal of Medicine. 1976;294(9):482-3.
  28. Davidson CS. The Metabolic Basis of Inherited Disease. Fifth Edition. Edited by John B. Stanbury, James B. Wyn-gaarden, Donald S. Fredrickson, Joseph L. Goldstein, and Michael S. Brown, xvi + 2,032 pp. New York: McGraw-Hill, 1983. Hepatology. 1983;3(3):461-2.
  29. Kalter H. The metabolic basis of inherited disease. 3rd Ed. J. B. Stanbury, J. B. Wyngaarden, and D. S. Fredrickson, eds. McGraw-Hill, New York. 1778 pp. 1972. Teratology. 1972;6(3):362-.
  30. Hubacek JA, Berge KE, Cohen JC, Hobbs HH. Mutations in ATP-cassette binding proteins G5 (ABCG5) and G8 (ABCG8) causing sitosterolemia. Hum Mutat. 2001;18(4):359-60.
  31. Watts GF, Mitchell WD. Clinical and metabolic findings in a patient with phytosterolaemia. Ann Clin Biochem. 1992;29 ( Pt 2):231-6.
  32. Hidaka H, Yasuda H, Kobayashi M, Hatanaka I, Takahashi M, Matsumoto K, et al. Familial spinal xanthomatosis with sitosterolemia. Intern Med. 1992;31(8):1038-42.
  33. Salen G, Shefer S, Nguyen L, Ness GC, Tint GS, Shore V. Sitosterolemia. J Lipid Res. 1992;33(7):945-55.
  34. Björkhem I, Boberg KM, Leitersdorf E. Inborn Errors in Bile Acid Biosynthesis and Storage of Sterols Other than Cholesterol. In: Valle D, Antonarakis S, Ballabio A, Beaudet A, Mitchell GA, editors. The Online Metabolic and Molecular Bases of Inherited Disease. New York, NY: McGraw-Hill Education; 2019.
  35. Salen G, Patel S, Batta AK. Sitosterolemia. Cardiovasc Drug Rev. 2002;20(4):255-70.
  36. Su Y, Wang Z, Yang H, Cao L, Liu F, Bai X, et al. Clinical and molecular genetic analysis of a family with sitosterolemia and co-existing erythrocyte and platelet abnormalities. Haematologica. 2006;91(10):1392-5.
  37. Wang G, Wang Z, Liang J, Cao L, Bai X, Ruan C. A phytosterolemia patient presenting exclusively with macrothrombocytopenia and stomatocytic hemolysis. Acta Haematol. 2011;126(2):95-8.
  38. Kaya Z, Niu DM, Yorulmaz A, Tekin A, Gursel T. A novel mutation of ABCG5 gene in a Turkish boy with phytosterolemia presenting with macrotrombocytopenia and stomatocytosis. Pediatr Blood Cancer. 2014;61(8):1457-9.
  39. Wang Z, Cao L, Su Y, Wang G, Wang R, Yu Z, et al. Specific macrothrombocytopenia/hemolytic anemia associated with sitosterolemia. Am J Hematol. 2014;89(3):320-4.
  40. Gülen H, Yıldırım AT, Yiğit Y, Yorulmaz A. Typical hematological findings facilitating the diagnosis of sitosterolemia. Pediatr Int. 2021;63(4):472-3.
  41. Gok V, Tada H, Ensar Dogan M, Alakus Sari U, Aslan K, Ozcan A, et al. A teenager boy with a novel variant of Sitosterolemia presented with pancytopenia. Clin Chim Acta. 2022;529:61-6.
  42. Mandato C, Siano MA, Nazzaro L, Gelzo M, Francalanci P, Rizzo F, et al. A ZFYVE19 gene mutation associated with neonatal cholestasis and cilia dysfunction: case report with a novel pathogenic variant. Orphanet J Rare Dis. 2021;16(1):179.
  43. Wang Y, Guo YL, Dong QT, Li JJ. Severe aortic valve stenosis in a 14-year-old boy with sitosterolemia. J Clin Lipidol. 2018.
  44. Xia Y, Duan Y, Zheng W, Liang L, Zhang H, Luo X, et al. Clinical, genetic profile and therapy evaluation of 55 children and 5 adults with sitosterolemia. J Clin Lipidol. 2022;16(1):40-51.
  45. Tada H, Kojima N, Yamagami K, Takamura M, Kawashiri MA. Clinical and genetic features of sitosterolemia in Japan. Clin Chim Acta. 2022;530:39-44.
  46. Kiss S, Lee JY, Pitt J, MacGregor D, Wallace J, Marty M, et al. Dig deeper when it does not make sense: Juvenile xanthomas due to sitosterolemia. JIMD Rep. 2020;56(1):34-9.
  47. Lee JH, Song DY, Jun SH, Song SH, Shin CH, Ki CS, et al. High prevalence of increased sitosterol levels in hypercholesterolemic children suggest underestimation of sitosterolemia incidence. PLoS One. 2020;15(8):e0238079.
  48. Tada H, Okada H, Nomura A, Yashiro S, Nohara A, Ishigaki Y, et al. Rare and Deleterious Mutations in ABCG5/ABCG8 Genes Contribute to Mimicking and Worsening of Familial Hypercholesterolemia Phenotype. Circ J. 2019;83(9):1917-24.
  49. Tada MT, Rocha VZ, Lima IR, Oliveira TGM, Chacra AP, Miname MH, et al. Screening of ABCG5 and ABCG8 Genes for Sitosterolemia in a Familial Hypercholesterolemia Cascade Screening Program. Circ Genom Precis Med. 2022;15(3):e003390.
  50. Nomura A, Emdin CA, Won HH, Peloso GM, Natarajan P, Ardissino D, et al. Heterozygous ATP-binding Cassette Transporter G5 Gene Deficiency and Risk of Coronary Artery Disease. Circulation: Genomic and Precision Medicine.2020;13(5):417-423..
  51. Alagille D, Odievre M, Gautier M, Dommergues JP. Hepatic ductular hypoplasia associated with characteristic facies, vertebral malformations, retarded physical, mental, and sexual development, and cardiac murmur. J Pediatr. 1975;86(1):63-71.
  52. Kojima N, Tada H, Usui S, Sakata K, Hayashi K, Nohara A, et al. Serum sitosterol level predicting ABCG5 or ABCG8 genetic mutations. Clin Chim Acta. 2020;507:11-6.
  53. Wu M, Pei Z, Sun W, Wu H, Sun Y, Wu B, et al. Age-related reference intervals for serum phytosterols in children by gas chromatography-mass spectrometry and its application in diagnosing sitosterolemia. Clin Chim Acta. 2023;540:117234.
  54. Brown EE, Sturm AC, Cuchel M, Braun LT, Duell PB, Underberg JA, et al. Genetic testing in dyslipidemia: A scientific statement from the National Lipid Association. J Clin Lipidol. 2020;14(4):398-413.
  55. Rios J, Stein E, Shendure J, Hobbs HH, Cohen JC. Identification by whole-genome resequencing of gene defect responsible for severe hypercholesterolemia. Hum Mol Genet. 2010;19(22):4313-8.
  56. Hamdan IJA, Sanchez-Siles LM, Garcia-Llatas G, Lagarda MJ. Sterols in Infant Formulas: A Bioaccessibility Study. Journal of Agricultural and Food Chemistry. 2018;66(6):1377-85.
  57. Llop JM, Virgili N, Moreno-Villares JM, Garcia-Peris P, Serrano T, Forga M, et al. Phytosterolemia in parenteral nutrition patients: implications for liver disease development. Nutrition. 2008;24(11-12):1145-52.
  58. Kratz M, Kannenberg F, Gramenz E, Berning B, Trautwein E, Assmann G, et al. Similar serum plant sterol responses of human subjects heterozygous for a mutation causing sitosterolemia and controls to diets enriched in plant sterols or stanols. Eur J Clin Nutr. 2007;61(7):896-905.
  59. Myrie SB, Mymin D, Triggs-Raine B, Jones PJ. Serum lipids, plant sterols, and cholesterol kinetic responses to plant sterol supplementation in phytosterolemia heterozygotes and control individuals. Am J Clin Nutr. 2012;95(4):837-44.
  60. Gregg RE, Connor WE, Lin DS, Brewer HB, Jr. Abnormal metabolism of shellfish sterols in a patient with sitosterolemia and xanthomatosis. The Journal of Clinical Investigation. 1986;77(6):1864-72.
  61. Parsons HG, Jamal R, Baylis B, Dias VC, Roncari D. A marked and sustained reduction in LDL sterols by diet and cholestyramine in beta-sitosterolemia. Clin Invest Med. 1995;18(5):389-400.
  62. Tsubakio-Yamamoto K, Nishida M, Nakagawa-Toyama Y, Masuda D, Ohama T, Yamashita S. Current Therapy for Patients with Sitosterolemia --Effect of Ezetimibe on Plant Sterol Metabolism. Journal of atherosclerosis and thrombosis. 2010;17(9):891-900..
  63. Cobb MM, Salen G, Tint GS. Comparative effect of dietary sitosterol on plasma sterols and cholesterol and bile acid synthesis in a sitosterolemic homozygote and heterozygote subject. J Am Coll Nutr. 1997;16(6):605-13.
  64. Nguyen LB, Cobb M, Shefer S, Salen G, Ness GC, Tint GS. Regulation of cholesterol biosynthesis in sitosterolemia: effects of lovastatin, cholestyramine, and dietary sterol restriction. J Lipid Res. 1991;32(12):1941-8.
  65. Connor WE, Lin DS, Pappu AS, Frohlich J, Gerhard G. Dietary sitostanol and campestanol: accumulation in the blood of humans with sitosterolemia and xanthomatosis and in rat tissues. Lipids. 2005;40(9):919-23.
  66. Othman RA, Myrie SB, Mymin D, Merkens LS, Roullet JB, Steiner RD, et al. Ezetimibe reduces plant sterol accumulation and favorably increases platelet count in sitosterolemia. J Pediatr. 2015;166(1):125-31.
  67. Salen G, von Bergmann K, Lutjohann D, Kwiterovich P, Kane J, Patel SB, et al. Ezetimibe effectively reduces plasma plant sterols in patients with sitosterolemia. Circulation. 2004;109(8):966-71.
  68. Lutjohann D, von Bergmann K, Sirah W, Macdonell G, Johnson-Levonas AO, Shah A, et al. Long-term efficacy and safety of ezetimibe 10 mg in patients with homozygous sitosterolemia: a 2-year, open-label extension study. Int J Clin Pract. 2008;62(10):1499-510.
  69. Salen G, Starc T, Sisk CM, Patel SB. Intestinal cholesterol absorption inhibitor ezetimibe added to cholestyramine for sitosterolemia and xanthomatosis. Gastroenterology. 2006;130(6):1853-7.
  70. Belamarich PF, Deckelbaum RJ, Starc TJ, Dobrin BE, Tint GS, Salen G. Response to diet and cholestyramine in a patient with sitosterolemia. Pediatrics. 1990;86(6):977-81.
  71. Tanaka H, Watanabe Y, Hirano S, Tada H, Nomura A, Kawashiri MA, et al. Sitosterolemia Exhibiting Severe Hypercholesterolemia with Tendon Xanthomas Due to Compound Heterozygous ABCG5 Gene Mutations Treated with Ezetimibe and Alirocumab. Intern Med. 2020;59(23):3033-7.
  72. Nguyen LB, Shefer S, Salen G, Horak I, Tint GS, McNamara DJ. The effect of abnormal plasma and cellular sterol content and composition on low density lipoprotein uptake and degradation by monocytes and lymphocytes in sitosterolemia with xanthomatosis. Metabolism. 1988;37(4):346-51.
  73. Nguyen LB, Shefer S, Salen G, Ness GC, Tint GS, Zaki FG, et al. A molecular defect in hepatic cholesterol biosynthesis in sitosterolemia with xanthomatosis. The Journal of Clinical Investigation. 1990;86(3):923-31.
  74. Salen G, Batta AK, Tint GS, Shefer S, Ness GC. Inverse relationship between plasma cholestanol concentrations and bile acid synthesis in sitosterolemia. J Lipid Res. 1994;35(10):1878-87.

 

Thyroid Hormone Serum Transport Proteins

ABSTRACT

 

Thyroid hormone (TH) effects are dependent on the quantity of the hormone that reaches the tissues, hormone activation, and the availability of unaltered TH receptors in the cell’s nuclei and cytoplasm. Since TH enters the cell unbound, the concentration of free rather than total hormone reflects more accurately the activity level of TH-dependent processes. Under normal conditions, changes in free hormone level are adjusted by appropriate stimulation or suppression of hormone secretion and disposal. Total TH concentration in serum is normally kept at a level proportional to the concentration of carrier proteins, and appropriate to maintain a constant free hormone level. 

 

INTRODUCTION

 

Most carrier protein dependent alterations in total hormone concentration in serum are due to quantitative changes in the hormone-binding proteins and less commonly to changes in affinities for the hormone.  Since wide fluctuations in the concentration of TH carrier proteins does not alter the hormonal economy or metabolic status of the subject (1), their function is open to speculation.  They are responsible for the maintenance of a large extrathyroidal pool of TH of which only the minute, <0.5 % fraction of free hormone is immediately available to tissues.  It can be estimated that in the absence of binding proteins the small extrathyroidal T4 pool would be significantly reduced, if not completely depleted in a matter of hours following a sudden cessation of hormone secretion.  In contrast, in the presence of normal concentrations of T4-binding serum proteins, and in particular thyroxine-binding globulin (TBG), a 24-h arrest in hormonal secretion would bring about a decrease in the concentration of T4 and T3 in the order of only 10 and 40 per cent, respectively.  Thus, it seems logical to assume that one of the functions of T4-binding proteins in serum is to safeguard the body from the effects of abrupt fluctuations in hormonal secretion.  The second likely function of T4-binding serum proteins is to serve as an additional protection against iodine wastage by imparting macromolecular properties to the small iodothyronine molecules, thus limiting their urinary loss (2).  The lack of high affinity T4-binding proteins in fish (3), for example, may be teleologically attributed to the greater iodine abundance in their natural habitat.  Liver perfusion studies suggest a third function, that facilitating the uniform cellular distribution of T4, allowing for changes in the circulating thyroid hormone level to be rapidly communicated to all cells within organ tissues (4).  A fourth function, modeled after the corticosteroid-binding globulin (5), is targeting the amount of hormone delivery by site specific, enzymatic, alteration of TBG.  Indeed neutrophil derived elastase transforms TBG into a heat resistant, relaxed, form with reduced T4-binding affinity (6).  TBG was found to have a putative role on the testicular size of the boar.  In fact, Meishan pigs with histidine rather than an asparagine in codon 226 have a TBG with lower affinity for T4, smaller testes and earlier onset of puberty (7, 8).

 

In normal man, approximately 0.03 per cent of the total serum T4, and 0.3 per cent of the total serum T3 are present in free or unbound form (3, 9).  The major serum thyroid hormone-binding proteins are thyroxine-binding globulin [TBG or thyropexin], transthyretin [TTR or thyroxine-binding prealbumin (TBPA)], and albumin (HSA, human serum albumin)(10).  Several other serum proteins, in particular high density lipoproteins, bind T4 and T3 as well as rT3 (9, 11) but their contribution to the overall hormone transport is negligible in both physiological and pathological situations.  In term of their relative abundance in serum, HSA is present at approximately 100-fold the molar concentration of TTR and 2,000-fold that of TBG.  However, from the view point of the association constants for T4, TBG has highest affinity, which is 50-fold higher than that of TTR and 7,000-fold higher that of HSA.  As a result, TBG binds 75% of serum T4, while TTR and HSA binds only 20% and 5%, respectively (Table 1).  The distribution of the iodothyronine metabolites among the three serum binding proteins is distinct (12). According to their affinity, T4 > tetraiodothyroacetic acid (TETRAC or T4A) = 3,3’,5’-triiodothyronine (reverse T3 or rT3) > T3 > triiodothyroacetic acid (TRIAC or T3A) = 3,3’-diiodothyronine (T2) > 3-monoipdothyronine (T1) = 3,5-T2 > thyronine (T0) for TBG (IC50-range: 0.36 nM to >100 lM) and T4A > T4 = T3A > rT3>T3 > 3,3’-T2 > 3-T1 > 3,5-T2 > T0 for transthyretin (IC50-range: 0.94 nM to >100 lM).  TBG, transthyretin, and albumin were not associated with T0, 3-T1, 3,3-T2, rT3, and T4A.  From evolutionary point of view, the three iodothyronine-binding serum proteins developed in reverse order of their affinity for T4, HSA being the oldest (13).

 

Table 1. Some Properties and Metabolic Parameters of the Principal TH-Binding Proteins in Serum

 

TBG

TTR

HSA

Molecular weight (K daltons)

54*

55

66.5

Structure

Monomer

Tetramer

Monomer

Carbohydrate content (%)

20

 

 

Number of binding sites for T4 and T3

1

2

4

Association constant, Ka (M-1)

 

 

 

       For T4

1 x 1010

2 x 108**

1.5 x 106**

       For T3

1 x 109

1 x 106

2 x 105

Concentration in serum

 

 

 

         (mean normal, mg/liter)

16

250

40,000

Relative distribution of T4 and T3 in serum (%)

 

 

 

       For T4

75

20

5

       For T3

75

<5

20

In-Vivo Survival

 

 

 

Half‑life (days)

5***

2

15

Degradation rate (mg/day)

15

650

17,000

 *Apparent molecular weight on acrylamide gel electrophoresis 60 K daltons.

 **Value given is for the high affinity binding site only.

***Longer under the influence of estrogen.

 

The existence of inherited TH-binding protein abnormalities was recognized 1959, with the report of a family with TBG-excess (14) but it took 30 years before the first mutation in the TBG (serine protease inhibitor, SERPIN A7) gene was identified (15).  Genetic variants of TH-binding proteins having different capacity or affinity for their ligands than the common type protein result in euthyroid hyper- or hypo-iodothyroninemia.  The techniques of molecular biology have traced these abnormalities to polymorphisms or mutations in genes encoding TBG and TTR and HSA (see Chapter on Defects of Thyroid Hormone Transport in Serum).

 

THYROXINE-BINDING GLOBULIN (TBG)

 

The Molecule, Structure and Physical Properties  

 

TBG is a 54 kD acidic glycoprotein migrating in the inter-α-globulin zone on conventional electrophoresis, at pH 8.6.  The term, thyroxine-binding globulin, is a misnomer since the molecule also binds T3 and reverse T3.  It was first recognized to serve as the major thyroid hormone transport protein in serum in 1952 (16).  Since TBG binds 75% of serum T4 and T3, quantitative and qualitative abnormalities of this protein have most profound effects on the total iodothyronine levels in serum.  Its primary structure was deduced in 1989 from the nucleotide sequence of a partial TBG cDNA and an overlapping genomic DNA clones (17).  However, it took 17 years to characterize its three dimensional structure by crystallographic analysis (18) (Fig. 1).

 

Figure 1. Structure of the TBG molecule: Reactive loop (in yellow). Insertion occurs following its cleavage by proteases to give an extra strand in the main sheet of the molecule but the T4-binding site can still retain its active conformation. This is in keeping with other findings showing that the binding and release of T4 is not due to a switch from an on to an off conformation but rather results from an equilibrated change in plasticity of the binding site. So, the S-to-R change in TBG results in a 6 -fold decrease but not a total loss of affinity. The important corollary is that that the release of thyroxine is a modulated process as notably seen in response to changes in temperature (19). (Courtesy of Dr, R.W. Carrell),

TBG is synthesized in the liver as single polypeptide chain of 415 amino acids.  The mature molecule, minus the signal peptide, is composed of 395 amino acids (44 kD) and four heterosaccharide units with 5 to 9 terminal sialic acids.  The carbohydrate chains are not required for hormone binding but are important for the correct post-translational folding and secretion of the molecule (20, 21) and are responsible for the multiple TBG isoforms (microheterogeneity) present on isoelectric focusing (22, 23).  The isoelectric point of normal TBG ranges from pH 4.2 to 4.6, however, this increases to 6 when all sialic acid residues are removed.

 

The protein is very stable when stored in serum, but rapidly loses its hormone binding properties by denaturation at temperatures above 55°C and pH below 4.  The half-life of denaturation at 60°C is approximately 7 min but association with T4 increases the stability of TBG (24-26).  TBG can be measured by immunometric techniques or saturation analysis using one of its iodothyronine ligands (26-28).

 

The tertiary structure of TBG was solved by co-crystallizing the in-vitro synthesized non-glycosylated molecule with T4 and speculations regarding the properties of TBG and its variants have been confirmed (18, 19). The molecule caries T4 in a surface pocket held by a series of hydrophobic interactions with underlying residues and hydrogen bonding of the aminoproprionate of T4 with adjacent residues (Figure 1). TBG differs from other members of the SERPIN family in having the upper half of the main ß-sheet completely opened. This allows the reactive center peptide loop to move in and out of the sheet, resulting in binding and release of the ligand without cleavage of TBG. Thus the molecule can assume a high-affinity and a low-affinity conformation, a model proposed earlier by Grasberger et al (29) and confirmed crystallographically (18). This reversibility is due to the unique presence of P8 proline in TBG, rather than a threonine in all other SERPINs, limiting loop insertion.  The coordinated movements of the reactive loop, hD, and the hormone-binding site allow the allosteric regulation of hormone release.

 

Gene Structure and Transcriptional Regulation

 

The molecule is encoded by a single gene copy located in the long arm of the human X-chromosome (Xq22.2) (30, 31).  The gene consists of 5 exons spanning 5.5kbp (Fig. 2).  The first exon is a small and non-coding.  It is preceded by a TATAA box and a sequence of 177 nucleotides containing an hepatocyte transcription factor-1 (HNF-1) binding motif that imparts to the gene a strong liver specific transcriptional activity (32).  The numbers and size of exons, their boundaries and types of intron splice junctions as well as the amino acid sequences they encode are similar to those of other members of the SERPIN family, to which TBG belongs (32).  These include cortisol-binding globulin and the serine protease inhibitors, α1-antitrypsin (α1AT) and α1-antichymotrypsin (α1ACT).

 

Figure 2. A. Genomic organization and chromosomal localization of thyroid hormone serum binding proteins. Filled boxes represent exons. Location of initiation codons and termination codons are indicated by arrows. B. Structure of promoter regions with the location of cis-acting transcriptional regulatory elements. Reproduced with permission from Hayashi and Refetoff, Molecular Endocrinology: Basic concepts and clinical correlations, Raven Press Ltd. 1995.

Biological Properties

 

The TBG molecule has a single iodothyronine binding site with affinity slightly higher for T4 than for T3 (33) (Table 1). Optimal binding activity requires the presence of the L-alanine side chain, an unsubstituted 4'-hydroxyl group, a diphenyl ether bridge, and halogen (I or Br) constituents at the 3,5,3' and 5' positions (34).  Compared to L-T4, 3,3’,5’-triiodothyronine (rT3) binds to TBG with ~40% higher affinity, D-T4 with half that of the L-isomer and tetraiodothyroacetic acid with ~25%.  A number of organic compounds compete with thyroid hormone-binding to TBG.  Most notable are: 5,5‑diphenylhydantoin (35), 1,8-anilinonaphthalenesulfonic acid, and salicylates (36).  While reversible flip-flop conformational changes of TBG allow for binding and release of the hormone ligand, cleavage of the molecule by leukocyte elastase produces a permanent change in the properties of the molecule.  This modified form has reduced T4-binding and increased heat stability (6).

 

Denatured TBG does not bind iodothyronines but can be detected with antibodies that recognize the primary structure of the molecule (26).  In euthyroid adults with normal TBG concentration, about one-third of the molecules carry thyroid hormone, mainly T4.  When fully saturated, it carries about 20 µg of T4/dl of serum.  The biologic half-life is about 5 days, and the volume of distribution is similar to that of albumin (37, 38) (Table 1).  TBG is cleared by the liver.  Loss of sialic acid accelerates its removal through interaction with the asialo-glycoprotein receptors reducing the half live by 500-fold (24).  However, it is unknown whether desialylation is a required in the normal pathway of TBG metabolism.

 

Physiology

 

TBG concentration in the serum of normal adults ranges from 1.1 to 2.1 mg/dl (180 - 350 nM), 14 - 26 µg T4/dl in terms of maximal T4-binding capacity.   The protein is present in serum of the 12th week old fetus and in the newborn until 2-3 years of age it is about 1.5 times the normal adult concentration (39-41).  TBG levels decline slightly reaching a nadir during mid-adulthood and tend to rise with further advance in age (42).  Variable amounts of TBG, though much smaller than those in serum, have been detected in amniotic fluid (43), cerebrospinal fluid (44) and urine (45).

 

Estrogen excess, either from an endogenous source (hydatidiform mole, estrogen-producing tumors, etc.) or exogenous (therapeutic or birth control use) is the most common cause of increased serum TBG concentration.  The level of several other serum proteins such as corticosteroid-binding globulin, testosterone-binding globulin, ceruloplasmin, and transferrin, are also increased (46).  This effect of estrogen is mediated through an increase in the complexity of the oligosaccharide residues in TBG together with an increase in the number of sialic acids resulting in prolonged biological half-life (47, 48).  Androgens and anabolic steroids produce an opposite effect (49, 50).  Although sex hormones affect the serum level of TBG, gender differences are small except during pregnancy during which concentrations are on the average 2.5-fold the normal value (28, 51).  Extreme changes in TBG concentration (low or high) alters the accuracy of immunometric measurements of free iodothyronines and particularly that of T3 (52).

 

Acquired TBG Abnormalities

 

Altered synthesis, degradation, or both are responsible for the majority of acquired TBG abnormalities (38).  Severe terminal illness is undoubtedly the most common cause for acquired decrease in TBG concentration.  Interleukin-6, a stimulator of acute phase reactants, is a candidate for mediation of this effect (53).  In vivo studies in man showed a reduction in the turnover of TBG in hypothyroidism and an increase in hyperthyroidism (37, 38).  Thus, alterations in the degradation rate, rather than changes in the rate of synthesis, may be responsible for the changes of TBG concentration observed in these two conditions.

 

Partially desialylated TBG, has slow electrophoretic mobility (sTBG, not to be confused with the variant TBG-S), and was found in the serum of some patients with severe liver disease (54) and may be present in relatively higher proportion than TBG in serum of patients with a variety of non-thyroidal illnesses and particularly those with compromised hepatocellular function (55).  This is not surprising considering that sTBG is removed by the asialoglycoprotein receptors present in abundance on liver cells (24, 56).

 

Patients with the carbohydrate-deficient glycoprotein (CDG) syndrome show a characteristic cathodal shift in the relative proportion of TBG isoforms compatible with diminished sialic acid content (57).  This inherited syndrome presenting psychomotor retardation, cerebellar hypoplasia, peripheral sensorimotor neuropathy, and variably, retinitis pigmentosa, skeletal abnormalities and lipodystrophy (58), manifests also abnormalities of charge and mass in a variety of serum glycoproteins (59).

 

TRANSTHYRETIN (TTR)

 

The Molecule, Structure and Physical Properties 

 

TTR is a 55kD homotetramer which is highly acidic although it contains no carbohydrate.  Formerly known as thyroxine-binding prealbumin (TBPA), for its electrophoretic mobility anodal to albumin, was first recognized to bind T4 in 1958 (60).  Subsequently it was demonstrated that TTR also forms a complex with retinol-binding protein and thus plays a role in the transport of vitamin A (retinol, or trans retinoic acid) (61, 62).

 

TTR circulates in blood as a stable tetramer of identical subunits, each containing 127 amino acids (63).  Although the tetrameric structure of the molecule was demonstrated by genetic studies (64, 65), detailed structural analysis is available through X-ray crystallography (66, 67) (Fig. 3).  Each TTR subunit has 8 ß-strands four of which form the inner sheet and four the outer sheet.  The four subunits form a symmetrical ß-barrel structure with a double trumpeted hydrophobic channel that traverses the molecule forming the two iodothyronine binding sites.  Despite the apparent identity of the two iodothyronine binding sites, TTR usually binds only one T4 molecule because the binding affinity of the second site is greatly reduced through a negative cooperative effect (69).  The TTR tetramer can bind four molecules of RBP that do not interfere with T4-binding, and vice versa (70).  TTR can be measured by densitometry after its separation from the other serum proteins by electrophoresis, by hormone saturation, and by immunoassays.

 

Figure 3. X-ray structure of TTR. The molecule is a homotetrameric protein composed of four monomers of 127 amino acids. Structurally, in its native state, TTR contains eight stands (A-H) and a small α-helix. The contacts between the dimers form two hydrophobic pockets where T4 binds (T4 channel). As shown in the magnified insert, each monomer contains one small α-helix and eight β-strands (CBEF and DAGH). Adapted from a model; PDB code 1DVQ (68).

Gene Structure and Transcriptional Regulation  

 

TTR is encoded by a single gene copy located on human chromosome 18 (18q11.2-12.1) (63, 71) (Fig. 2).  The gene consists of 4 exons spanning for 6.8kbp.  Knowledge about the transcriptional regulation of the human TTR gene comes from studies of the mouse gene structural and sequence homology which extends to the promoter region (72, 73).  In both species a TATAA box and binding sites for HNF-1, 3 and 4 are located within 150 bp from the transcription start site.

 

Although TTR in serum originates from the liver (74), TTR mRNA is also found in kidney cells, the choroid plexus, meninges, retina, placenta, pancreatic islet cells and fetal intestine (75-78).  TTR constitutes up to 25% of the total protein present in ventricular cerebrospinal fluid where it binds 80% of T4 (79).

 

Biological Properties

 

Despite the 20-fold higher concentration of TTR in serum relative to that of TBG, it plays a lesser role in iodothyronine transport.  In the presence of normal levels of TBG, wide fluctuations in TTR concentration or its removal from serum by specific antibodies has little influence on the concentration of free T4 (80).  Some of the properties of TTR are summarized in Table 1.

 

The first T4 molecule binds to TTR with a Ka of about 100‑fold higher than that for HSA and about 100-fold lesser than that for TBG.  Properties necessary for optimal binding activity include iodines at the 3' and 5' positions and a desamino acid side chain which explain the lower T3 and higher T4A  affinities relative to that of T4 (34, 81).  Non-iodothyronine ligands are also differentially bound, the most notable example being the flavonoid compounds which have a markedly higher binding affinity for TTR than for TBG (82).  Among drugs that compete with T4-binding to TTR are ethacrynic acid, salicylates, 2,4-dinitrophenol, penicillin (83, 84) and perfluoroalkyl substances (85).  The latter have with near equal affinity to TTR and TBG.  Barbital also inhibits iodothyronine binding to TTR.

 

Only 0.5% of the circulating TTR is occupied by T4.  TTR has a relatively rapid turnover (t1/2 = 2 days) and a distribution space similar to that of HSA and TBG (86, 87) except that it also exists in CSF. Hence, acute diminution in the rate of synthesis is accompanied by a rapid decrease of its concentration in serum.

 

Physiology 

 

Normal average concentration in serum is 25 mg/dl, and corresponds to a maximal binding capacity of approximately 300 µg T4/dl.  Changes in TTR concentration have relatively little effect on the serum concentration of serum iodothyronines (80, 88).  There is a distinct reciprocal relationship between acquired changes in TBG and TTR concentration related to gender, age, glucocorticoids, estrogen and androgens (42, 51, 89-91).

 

Acquired TTR Abnormalities 

 

The reduction or serum TTR concentration surpasses that of TBG in major illness, nephrotic syndrome, liver disease, cystic fibrosis, hyperthyroidism, and protein-calorie malnutrition (10, 92-94).  Increased serum TTR concentration can occur in some patients with islet cell carcinoma (95).  Studies on the metabolism of TTR in man, utilizing radioiodinated purified human TTR, indicate that diminished TTR concentration associated with severe illness or stress is due to a decrease in the rate of synthesis or an increase in the rate of degradation, or both (86, 87).

 

HUMAN SERUM ALBUMIN (HSA)

 

The Molecule, Structure and Physical Properties

 

HSA is a 66.5 kD protein synthesized by the liver.  It is composed of 585 amino acids with high content of cystines and charged amino acids but no carbohydrate (96).  The three domains of the molecule can be conceived as three tennis balls packaged in a cylindrical case.

 

Gene Structure and Transcriptional Regulation  

 

HSA is encoded by a single gene copy located on human chromosome 4 (4q11-q13) (97).  The gene contains 15 exons, 14 of which are coding (98) (Fig. 2). The promoter region of the HSA gene has been most intensive studied.  The transcriptional regulation has been best characterized in rodents that share 90% sequence homology with the corresponding human gene, including a distal enhancer element 10 kbp upstream from the promoter region (99).  Binding sites for hepatocyte enriched nuclear proteins, such as HNF-1, C/EBP, and DBP have been identified (100-102).

 

Biological Properties  

 

HSA associates with a wide variety of substances including hormones and drugs possessing a hydrophobic region, and thus the association of TH to HSA can be viewed as nonspecific.  Of the several iodothyronine-binding sites on the HSA molecule, only one has a relatively high affinity for T4 and T3.  Yet these are 10,000-fold inferior to those of TBG (27).  Fatty acids and chloride ions decrease their binding to HSA (27).  The biologic t1/2 of HSA is relatively long (103).  Some of its properties are summarized in Table 1.

 

More than half of the total protein content in serum is HSA.  As a result, it is the principal contributor to the maintenance of the colloid osmotic pressure (96).  It has been suggested that HSA synthesis may be, in part, regulated by a feedback mechanism involving alteration in the colloid osmotic pressure.  Indeed, down-regulation of HSA gene expression has been recently observed during the infusion of macromolecules in the rat (104).

 

Physiology 

 

Because of the low affinity and despite the high capacity of HSA for iodothyronines, its contribution to thyroid hormone transport is relatively minor.  Thus, even the most marked fluctuations of serum HSA concentration, including analbuminemia, have no significant effects on thyroid hormone levels (105).

 

LIPOPROTEINS

 

Lipoproteins bind T4, and to some extent T3 (9, 106). The affinity for T4-binding is similar to that of TTR.  These proteins are estimated to transport roughly 3% of the total T4 and perhaps as much as 6% of the total T3 in serum. The binding site of apolipoprotein A1 is a region of the molecule that is distinct from that portion which binds to the cellular lipoprotein receptors, and the physiological role of such binding is still unclear.

 

ACKNOWLEDGMENTS

 

Supported in part by grants DK-15070 from the National Institutes of Health (USA).

 

REFERENCES

 

  1. Refetoff S 1989 Inherited thyroxine-binding globulin (TBG) abnormalities in man. Endocr Rev 10:275-293.
  2. Chan V, Besser GM, Landon J 1972 Effects of oestrogen on urinary thyroxine excretion. Brit Med J 4:699-701.
  3. Refetoff S, Robin NI, Fang VS 1970 Parameters of thyroid function in serum of 16 selected vertebrate species: A study of PBI, serum T4, free T4, and the pattern of T4 and T3 binding to serum proteins. Endocrinology 86:793-805.
  4. Mendel CM, Weisinger RA, Jones AL, Cavalieri RR 1987 Thyroid hormone-binding proteins in plasma facilitate uniform distribution of thyroxine within tissues: A perfused rat liver study. Endocrinology 120:1742-1749.
  5. Pemberton PA, Stein PE, Pepys MB, Potter JM, Carell RW 1988 Hormone binding globulins undergo serpin conformational change in inflammation. Nature 336:257-258.
  6. Janssen OE, Golcher HMB, Grasberger H, Saller B, Mann K, Refetoff S 2002 Characterization of thyroxine-binding globulin cleaved by human leukocyte elastase. J Clin Endocrinol Metab 87:1217-1222.
  7. Ford JJ, Rohrer GA, Nonneman DJ, Lunstra DD, Wise TH 2010 Association of allelic variants of thyroid-binding globulin with puberty in boars and responses to hemicastration. Anim Reprod Sci 119:228-234.
  8. Nonneman D, Rohrer GA, Wise TH, Lunstra DD, Ford JJ 2005 A variant of porcine thyroxine-binding globulin has reduced affinity for thyroxine and is associated with testis size. Biol Reprod 72:214-220.
  9. Freeman T, Pearson JD 1969 The use of quantitative immunoelectrophoresis to investigate thyroxine-binding human serum proteins. Clin Chim Acta 26:365-368.
  10. Oppenheimer JH 1968 Role of plasma proteins in the binding, distribution, and metabolism of the thyroid hormones. N Engl J Med 278:1153-1162.
  11. Benvenga S, Gregg RE, Robbins J 1988 Binding of thyroid hormone to human plasma lipoproteins. J Clin Endocrinol Metab 67:6-16.
  12. Jongejan RMS, Meima ME, Visser WE, Korevaar TIM, van den Berg SAA, Peeters RP, de Rijke YB 2022 Binding Characteristics of Thyroid Hormone Distributor Proteins to Thyroid Hormone Metabolites. Thyroid.
  13. Yamauchi K, Ishihara A 2009 Evolutionary changes to transthyretin: developmentally regulated and tissue-specific gene expression. FEBS J 276:5357-5366.
  14. Beierwaltes WH, Robbins J 1959 Familial increase in the thyroxine-binding sites in serum alpha globulin. J Clin Invest 38:1683-1688.
  15. Mori Y, Seino S, Takeda K, Flink IL, Murata Y, Bell GI, Refetoff S 1989 A mutation causing reduced biological activity and stability of thyroxine-binding globulin probably as a result of abnormal glycosylation of the molecule. Mol Endocrinol 3:575-579.
  16. Gordon AH, Gross J, O'Connor D, Pitt-Rivers R 1952 Nature of circulating thyroid hormone-plasma protein complex. Nature 169:19-20.
  17. Flink IL, Bailey TJ, Gustefson TA, Markham BE, Morkin E 1986 Complete amino acid sequence of human thyroxine-binding globulin deduced from cloned DNA:Close homology to the serine antiproteases. Proc Natl Acad Sci U S A 83:7708-7712.
  18. Zhou A, Wei Z, Read RJ, Carrell RW 2006 Structural mechanism for the carriage and release of thyroxine in the blood. Proc Natl Acad Sci U S A 103:13321-13326.
  19. Qi X, Loiseau F, Chan WL, Yan Y, Wei Z, Milroy LG, Myers RM, Ley SV, Read RJ, Carrell RW, Zhou A 2011 Allosteric modulation of hormone release from thyroxine and corticosteroid-binding globulins. J Biol Chem 286:16163-16173.
  20. Murata Y, Magner JA, Refetoff S 1986 The role of glycosylation in the molecular conformation and secretion of thyroxine-binding globulin. Endocrinology 118:1614-1621.
  21. Kambe F, Seo H, Mori Y, Murata Y, Janssen OE, Refetoff S, Matsui N 1992 An additional carbohydrate chain in the variant thyroxine-binding globulin-Gary (TBGAsn-96) impairs its secretion. Mol Endocrinol 6:443-449.
  22. Gärtner R, Henze R, Horn K, Pickardt CR, Scriba PC 1981 Thyroxine-binding globulin: Investigation of microheterogeneity. J Clin Endocrinol Metab 52:657-664.
  23. Robbins J 1992 Thyroxine transport and the free hormone hypothesis. Endocrinology 131:546-547.
  24. Refetoff S, Fang VS, Marshall JS 1975 Studies on human thyroxine-binding globulin (TBG): IX. Some physical, chemical and biological properties of radioiodinated TBG and partially desialylated TBG (STBG). J Clin Invest 56:177-187.
  25. Grimaldi S, Edelhoch H, Robbins J 1982 Effects of thyroxine binding on the stability, conformation, and fluorescence properties of thyroxine-binding globulin. Biochemistry (Mosc) 21:145-150.
  26. Refetoff S, Murata Y, Vassart G, Chandramouli V, Marshall JS 1984 Radioimmunoassays specific for the tertiary and primary structures of thyroxine-binding globulin (TBG): Measurement of denatured TBG in serum. J Clin Endocrinol Metab 59:269-277.
  27. Tabachnick M, Giorgio NA, Jr. 1964 Thyroxine-protein interactions.II. The binding of thyroxine and its analogues to human serum albumin. Arch Biochem Biophys 105:563-569.
  28. Refetoff S, Hagen S, Selenkow HA 1972 Estimation of the T4 binding capacity of serum TBG and TBPA by a single T4 load ion exchange resin method. J Nucl Med 13:2-12.
  29. Grasberger H, Golcher HM, Fingerhut A, Janssen OE 2002 Loop variants of the serpin thyroxine-binding globulin: implications for hormone release upon limited proteolysis. Biochem J 365:311-316.
  30. Trent JM, Flink IL, Morkin E, Van TuinenP, Ledbetter  DH 1987 Localization of the human thyroxine-binding globulin gene to the long arm of the X chromosome (Xq21-22). Am J Hum Genet 41:428-435.
  31. Mori Y, Miura Y, Oiso Y, Seo H, Takazumi K 1995 Precise localization of the human thyroxine-binding globulin gene to chromosome Xq22.2 by fluorescence in situ hybridization. Hum Genet 96:481-482.
  32. Hayashi Y, Mori Y, Janssen OE, Sunthornthepvarakul T, Weiss RE, Takeda K, Weinberg M, Seo H, Bell GI, Refetoff S 1993 Human thyroxine-binding globulin gene: Complete sequence and transcriptional regulation. Mol Endocrinol 7:1049-1060.
  33. Hocman G 1981 Human thyroxine binding globulin. Rev Physiol Biochem Pharmacol 81:45-88.
  34. Cody V 1980 Thyroid hormone interactions: Molecular conformation, protein binding and hormone action. Endocr Rev 1:140-166.
  35. Oppenheimer JH, Tavernetti RR 1962 Displacement of thyroxine from human thyroxine-binding globulin by analogues of hydantoin.Steric aspects of the thyroxine-binding site. J Clin Invest 41:2213-2220.
  36. Larsen PR 1972 Salicylate-induced increases in free triiodothyronine in human serum: Evidence of inhibition of triiodothyronine binding to thyroxine-binding gloublin and thyroxine-binding prealbumin. J Clin Invest 51:1125-1134.
  37. Cavalieri RR, McMahon FA, Castle JN 1975 Preparation of 125I-labeled human thyroxine-binding alpha globulin and its turnover in normal and hypothyroid subjects. J Clin Invest 56:79-87.
  38. Refetoff S, Fang VS, Marshall JS, Robin NI 1976 Metabolism of thyroxine-binding globulin (TBG) in man:Abnormal rate of synthesis in inherited TBG deficiency and excess. J Clin Invest 57:485-495.
  39. Andreoli M, Robbins J 1962 Serum proteins and thyroxine-protein interaction in early human fetuses. J Clin Invest 41:1070-1077.
  40. Robbins J, Nelson JH 1958 Thyroxine-binding by serum protein in pregnancy and in the newborn. J Clin Invest 37:153-159.
  41. Stubbe P, Gatz J, Heidemann P, Muhlen ARG, Hesch R 1978 Thyroxine-binding globulin, triiodothyronine, thyroxine and thyrotropin in newborn infants and children. Horm Metab Res 10:58-61.
  42. Braverman LE, Dawber NA, Ingbar SH 1966 Observations concerning the binding of thyroid hormones in sera of normal subjects of varying ages. J Clin Invest 45:1273-1279.
  43. Burman KD, Read J, Dimond RC, Strum D, Wright FD, Patow W, Earll JM, Wartofsky L 1976 Measurement of 3,3',5'-Triiodothyroinine (reverse T3), 3,3'-L-diiodothyronine, T3 and T4 in human amniotic fluid and in cord and maternal serum. J Clin Endocrinol Metab 43:1351-1359.
  44. Hagen GA, Elliott WJ 1973 Transport of thyroid hormones in serum and cerebrospinal fluid. J Clin Endocrinol Metab 37:415-422.
  45. Gavin LA, McMahon FA, Castle JN, Cavalieri RR 1979 Detection of a thyroxine-binding protein physicochemically similar to serum thyroxine-binding globulin in normal human urine. J Clin Endocrinol Metab 48:843-847.
  46. Doe RP, Mellinger GT, Swaim WR, Seal JS 1967 Estrogen dosage effects on serum proteins: A longitudinal study. J Clin Endocrinol Metab 27:1081-1086.
  47. Ain KB, Mori Y, Refetoff S 1987 Reduced clearance rate of thyroxine-binding globulin (TBG) with increased sialylation: A mechanism for estrogen induced elevation of serum TBG concentration. J Clin Endocrinol Metab 65:689-696.
  48. Ain KB, Refetoff S, Sarne DH, Murata Y 1988 Effect of estrogen on the synthesis and secretion of thyroxine-binding globulin (TBG) by a human hepatoma cell line, Hep G2. Mol Endocrinol 2:313-323.
  49. Federman DD, Robbins J, Rall JE 1958 Effects of methyl testosterone on thyroid function, thyroxine metabolism, and thyroxine-binding protein. J Clin Invest 37:1024-1030.
  50. Barbosa J, Seal US, Doe RP 1971 Effects of anabolic steroids on haptoglobin, orosomucoid, plasminogen, fibrinogen, transferrin, ceruloplasmin, alpha-1-antitrypsin, beta- glucuronidase and total serum proteins. J Clin Endocrinol Metab 33:388-398.
  51. Braverman LE, Foster AE, Ingbar SH 1967 Sex-related differences in the binding in serumof thyroid hormone. J Clin Endocrinol Metab 27:227-232.
  52. Gant Kanegusuku A, Araque KA, Nguyen H, Wei B, Hosseini S, Soldin SJ 2021 The effect of specific binding proteins on immunoassay measurements of total and free thyroid hormones and cortisol. Ther Adv Endocrinol Metab 12:2042018821989240.
  53. Bartalena L, Farsetti A, Flink IL, Robbins J 1992 Effects of interleukin-6 on the expression of thyroid-hormone binding protein genes in cultured hepatoblastoma-derived (Hep G2) cells. Mol Endocrinol 6:935-942.
  54. Marshall JS, Pensky J, Green AM 1972 Studies on human thyroxine-binding globulin.IV. The nature of slow thyroxine-binding globulin. J Clin Invest 51:3173-3181.
  55. Reilly CP, Wellby ML 1983 Slow thyroxine binding globulin in the pathogenesis of increased dialysable fraction of thyroxine in nonthyroidal illnesses. J Clin Endocrinol Metab 57:15-18.
  56. Marshall JS, Green AM, Pensky J, Williams S, Zinn A, Carlson DM 1974 Measurement of circulating desialylated glycoproteins and correlation with hepatocellular damage. J Clin Invest 54:555-562.
  57. Macchia PE, Harrison HH, Scherberg NH, Sunthornthepvarakul T, Jaeken J, Refetoff S 1995 Thyroid function tests and characterization of thyroxine-binding globulin in the carbohydrate-deficient glycoprotein syndrome type I. J Clin Endocrinol Metab 80:3744-3749.
  58. Jaeken J, Hagberg B, Strømme P 1991 Clinical presentation and natural course of the carbohydrate-deficient glycoprotein syndrome. Acta Pediatr Scand Suppl 375:6-13.
  59. Stibler H, Jaeken J, Kristiansson B 1991 Biochemical characteristics and diagnosis of the carbohydrate-deficient glycoprotein syndrome. Acta Pediatr Scand Suppl 375:21-31.
  60. Ingbar SH 1958 A thyroxine-binding protein of human plasma. Endocrinology 63:256-259.
  61. Kanai M, Raz A, Goodman D 1968 Retinol-binding protein: The transport protein for vitamin A in human plasma. J Clin Invest 47:2025-2044.
  62. Peterson PA 1971 Characteristics of a vitamin A-transporting protein complex occurring in human serum. J Biol Chem 246:34-43.
  63. Tsuzuki T, Mita S, Maeda S, Araki S, Shimada K 1985 Stucture of human prealbumin gene. J Biol Chem260:12224-12227.
  64. Alper CA, Robin NI, Refetoff S 1969 Genetic polymorphism of Rhesus thyroxine-binding prealbumin: Evidence for tetrameric structure in primates. Proc Natl Acad Sci U S A 63:775-781.
  65. Bernstein RS, Robbins J, Rall JE 1970 Polymorphism of monkey thyroxine-binding prealbumin (TBPA): Mode of inheritance and hybridization. Endocrinol 86:383-390.
  66. Rerat C, Schwick HG 1967 Données cristallographiques sur la préalbumine du plasma sanguin. Acta Cryst 22:441.
  67. Blake CCF, Oatley SJ 1977 Protein-DNA and protein-hormone interactions in prealbumin: A model of the thyroid hormone nuclear receptor? Nature (London) 268:115-120.
  68. Azevedo E, Silva PF, Palhano F, Braga CA, Fouel D 2013 Transthyretin-related amyloidoses: a structural and thermodynamic approach. In: Feng D, (ed) Amyloidosis. Vol 1. InTech under CC BY 3.0 license.
  69. Irace G, Edelhoch H 1978 Thyroxine induced conformational changes in prealbumin. Biochemistry (Mosc) 17:5729-5733.
  70. van Jaarsveld PP, Edelhoch H, Goodman DS, Robbins J 1973 The interaction of human plasma retinol binding protein with prealbumin. J Biol Chem 248:4698-4705.
  71. LeBeau MM, Geurts van Kessel G 1991 Report of the committee on the genetic costitution of chromosome 18. Cytogenet Cell Genet 58:739-750.
  72. Costa RH, Grayson DR, Darnell Jr JE 1989 Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and a1-antitrypsin. Mol Cell Biol 9:1415-1425.
  73. Sladeck FM, Zhong R, Lai E, Darnell Jr. JE 1990 Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily. Gene Dev 4:2353-2365.
  74. Knowles BB, Howe CC, Aden DP 1980 Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen. Science 209:497-499.
  75. Dickson PW, ., Howlett GJ, Schreiber G 1985 Rat transthyretin (prealbumin):molecular cloning, nucleotide sequence, and gene expression in liver and brain. J Biol Chem 260:8214-.
  76. Dickson PW, Aldred AR, Marley PD, Bannister D 1985 Rat choroid plexus specializes in the synthesis and secretion of transthyretin (prealbumin). J Biol Chem 261:3475-.
  77. Jacobsson B, Pettersson T, Sandstedt B, Carlstrom A 1979 Prealbumin in the islets of Langerhans. Int Res Commun Syst Med Sci 7:590-.
  78. Richardson SJ 2009 Evolutionary changes to transthyretin: evolution of transthyretin biosynthesis. FEBS J 276:5342-5356.
  79. Herbert J, Wilcox JN, Pham K-TC, Fremeau Jr RT, Zeviani M, Dwork A, Soprano DR, Makover A, Goodman DS, Zimmerman EA, Roberts JL, Schon EA 1986 Transthyretin: a choroid plexus-specific transport protein in human brain. Neurology 36:900-911.
  80. Woeber KA, Ingbar SH 1968 The contribution of thyroxine-binding prealbumin to the binding of thyroxine in human serum, as assessed by immunoadsorption. J Clin Invest 47:1710-1721.
  81. Pages RA, Robbins J, Edelhoch H 1973 Binding of thyroxine and thyroxine analogs to human serum prealbumin. Biochem 12:2773-2779.
  82. Lueprasitsakul W, Alex S, Fang SL, Pino S 1990 Flavonoid administration immediately displaces thyroxine (T4) from serum transthyretin, increases serum free T4, and decreases thyrotropin in the rat. Endocrinology 126:2890-.
  83. Wolff J, Standaert ME, Rall JE 1961 Thyroxine displacement from serum proteins and depression of serum protein-bound iodine by certain drugs. J Clin Invest 40:1373-1379.
  84. Munro SL, Lim CF, Hall JG, Barlow JW, Craik DJ, Topliss DJ, Stockigt JR 1989 Drug competition for thyroxine binding to transthyretin (Prealbumin): Comparison with effects on thyroxine-binding globulin. J Clin Endocrinol Metab 68:1141-1147.
  85. Ren XM, Qin WP, Cao LY, Zhang J, Yang Y, Wan B, Guo LH 2016 Binding interactions of perfluoroalkyl substances with thyroid hormone transport proteins and potential toxicological implications. Toxicology 366-367:32-42.
  86. Socolow EL, Woeber KA, Purdy RH, Holloway MT, Ingbar SH 1965 Preparation of I131-labeled human serum prealbumin and its metabolism in normal and sick patients. J Clin Invest 44:1600.
  87. Oppenheimer JH, Surks MI, Bernstein G, Smith JC 1965 Metabolism of iodine-131-labeled thyroxine-binding prealbumin in man. Science 149:748-751.
  88. Braverman LE, AvRuskin T, Cullen MJ, Vagenakis AG, Ingbar SH 1971 Effects of norethandrolone on the transport and peripheral metabolism of thyroxine in patients lacking thyroxine-binding globulin. J Clin Invest 50:1644-1649.
  89. Braverman LE, Ingbar SH 1967 Effects of norethandrolone on the transport in serum and peripheral turnover of thyroxine. J Clin Endocrinol Metab 27:389-396.
  90. Oppenheimer JH, Werner SC 1966 Effect of prednisone on thyroxine-binding proteins. J Clin Endocrinol Metab 26:715-721.
  91. Man EB, Reid WA, Hellegers AE, Jones WS 1969 Thyroid function in human pregnancy. III. Serum thyroxine-binding prealbumin (TBPA) and thyroxine-binding globulin (TBG) of pregnant women aged 14 through 43 years. Am J Obst Gyn 103:338-347.
  92. Inada M, Sterling K 1967 Thyroxine turnover and transport in Laennec's cirrhosis of the liver. J Clin Invest 46:1275-1282.
  93. Ingenbleek Y, deVisscher M, deNayer P 1972 Measurement of prealbumin as index of protein-calorie malnutrition. Lancet 2:106-108.
  94. Smith FR, Underwood BA, Denning CR, Varma A, Goodman DSC 1972 Depressed plasma retinol-binding protein levels in cystic fibrosis. J Lab Clin Med 80:423-433.
  95. Rajatanavin R, Liberman C, Lawrence GD, D'Arcangues CM, Young RA, Emerson CH 1985 Euthyroid hyperthyroxinemia and thyroxine-binding prealbumin excess in islet cell carcinoma. J Clin Endocrinol Metab 61:17-21.
  96. Peters T, Jr. 1985 Serum albumin. Adv Prot Chem 37:161-245.
  97. Murray JC, Van Ommen GJB 1991 Report of the committee on the genetic constitution of chromosome 4. Cytogenet Cell Genet 58:231-260.
  98. Dugaiczyk A, Law SW, Dennison OE 1982 Nucleotide sequence and the encoded amino acids of human serum albumin mRNA. Proc Natl Acad Sci U S A 79:71-75.
  99. Hayashi Y, Chan J, Nakabayashi H, Hashimoto T, Tamaoki T 1992 Identification and characterization of two enhancers of the human albumin gene. J Biol Chem 267:14580-14585.
  100. Gereghini S, Raymondjean M, Carranca AG, Heibomel P, Yaniv M 1987 Factors involved in control of tissue-specific expression of albumin gene. Cell 50:627-638.
  101. Johnson PF 1990 Transcriptional activators in hepatocytes. Cell Growth Differ 1:47-52.
  102. Rey-Campos J, Yaniv M 1992 Regulation of albumin gene expression. Elsevier Science Publishers, Amsterdam.
  103. Beeken WL, Volwilier W, Goldsworthy PD, Garby LE, Reynolds WE, Stogsdill R, Stemler RS 1962 Studies of I131-albumin catabolism and distribution in normal young male adults. J Clin Invest 41:1312-1333.
  104. Pietrangelo A, Panduro A, Chowdhury JR, Shafritz DA 1992 Albumin gene expression is down-regulated by albumin or macromolecule infusion in the rat. J Clin Invest 89:1755-1760.
  105. Hollander CS, Bernstein G, Oppenheimer JH 1968 Abnormalities of thyroxine binding in analbuminemia. J Clin Endocrinol Metab 28:1064-1066.
  106. Benvenga S, Robbins J 1993 Lipoprotein-thyroid hormone interactions. TEM 4:194-198.

Cerebrotendinous Xanthomatosis

ABSTRACT

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder caused by homozygous or compound heterozygous mutations in the CYP27A1 gene. These patients lack mitochondrial sterol 27-hydroxylase enzyme, which is responsible for conversion of cholesterol to cholic acid and chenodeoxycholic acid (CDCA) in bile acid synthesis pathway. CYP27A1 mutation leads to decreased synthesis of bile acid, excess production of cholestanol, and consequent accumulation of cholestanol in tissues, including brain, leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts. The clinical manifestations usually start at infancy and develop during the first and second decades of life. The diagnosis of CTX is based on clinical findings, biochemical testing, and neuroimaging. Molecular genetic analysis although not necessary for initiation of treatment, provides definitive confirmation of CTX. Early initiation of CDCA is the treatment of choice for neurological and non-neurological symptoms of CTX and treatment with cholic acid has also been shown to be effective for non-neurological symptoms.

 

INTRODUCTION

 

Cerebrotendinous xanthomatosis (CTX; OMIM#213700) is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. It is caused by homozygous or compound heterozygous mutation in the CYP27A1 gene located on chromosome 2q33-qter. CYP27A1 gene encodes sterol 27-hydroxylase which is involved in bile acid synthesis. Over 100 different mutations (missense, deletions, insertions, splice site, and nonsense mutations) of the CYP27A1 gene have been reported worldwide in patients of different ethnic origin (1,2). About 50% of these mutations were found in the region of exons 6–8 of the CYP27A1 gene, however here is no genotype phenotype correlation (3,4). The metabolic pathway for bile acids synthesis is shown in the figure 1.

 

Figure 1. Bile acids are synthesized from cholesterol in the liver through two pathways: the classic pathway and the alternative pathway. The bile acid synthesis mainly produces two primary bile acids, cholic acid and chenodeoxycholic acid (CDCA). In the classic pathway, the mitochondrial sterol 27-hydroxylase (CYP27A1) catalyzes the steroid side-chain oxidation in both CA and CDCA synthesis. In the alternative pathway, CYP27A1 catalyzes the first step to convert cholesterol to 27-hydroxycholesterol which eventually is converted to CDCA.

 

CTX was first described by Van Bogart, Scherer and Epstein in 1937 as a condition characterized by early development of cataracts and large tendon xanthomas and later by progressive neurologic impairment (5,6). The incidence of CTX is estimated to be 3 to 5 per 100,000 people (1,7). The prevalence is estimated to be highest in Asians (1:44,407-93,084), intermediate in Europeans, Americans, and Africans/African Americans (1:70,795-233,597) and lowest in the Finnish population (1:3,388,767) (8). 

 

ETIOLOGY AND PATHOGENESIS

 

Patients with CTX lack mitochondrial sterol 27-hydroxylase, which is an important enzyme in both the alternative and classic bile acid synthesis pathways (9). The biochemical defect prevents the synthesis of chenodeoxycholic acid (CDCA) and cholic acid (10).The absence of the negative feedback mechanism of CDCA on 7a-hydroxylase, the rate limiting step, leads to the accumulation of cholestanol and its precursor 7α-hydroxy-4-cholesten-3-one (Figure 1) (9-11). Bile acid alcohols are glucoronidated and can be found in significantly increased amounts in blood, urine, and feces of untreated CTX subjects. It has been hypothesized that the increased bile alcohol levels may lead to disruption of the blood–brain barrier (BBB), and that increased permeability of BBB may be caused by circulating bile alcohol glucuronides (12). Cholestanol and cholesterol accumulate in many tissues, including brain (primarily white matter), leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts (13).

 

CLINICAL FEATURES

 

Patients with CTX present diverse manifestations with multi-organ involvement and a broad range of neurological and non-neurological symptoms (Table 1). The clinical manifestations usually start at infancy and develop during the first and second decades of life. Neonatal cholestatic jaundice and infantile-onset diarrhea with failure to thrive may be the earliest clinical manifestation (14-16). Childhood-onset cataracts are a common early symptom, described in 92% patients with CTX. Cataracts precede neurological signs and tendon xanthoma, and if present, are considered useful for early diagnosis (14,16,17). Tendon xanthomas have been documented in 71% patients with CTX and can appear in first, second or third decade. They are common in Achilles tendon, but can also be seen in fingers, tibial tuberosity, and triceps (17,18). Pes cavus deformity has also been described (19,20). Neurological dysfunction is almost always present in patients with CTX and usually occurs in late adolescents and early adulthood. The range of symptoms are wide and include intellectual disability; dementia; psychiatric symptoms (i.e., behavioral changes, depression, agitation, hallucination, and suicide attempts); pyramidal signs (spasticity, hyperreflexia, extensor plantar responses); cerebellar signs (progressive ataxia, dysarthria); movement disorder (parkinsonism, dystonia, myoclonus, postural tremor); seizures; and peripheral neuropathy (1,4,21-23). Osteoporosis, heart involvement, and premature atherosclerosis have also been described later in life (19,24-26).

 

Table 1. Clinical Features of Cerebrotendinous Xanthomatosis

Early childhood

            Neonatal cholestatic jaundice

            Chronic diarrhea

            Cataracts

            Developmental Delay

Late childhood

            Tendon xanthomas

            Psychiatric disorders

Adulthood

            Neurological dysfunction

                        Pyramidal signs (spasticity, hyperreflexia, extensor plantar responses)

                        Cerebellar signs (ataxia, dysarthria)

                        Movement disorder (parkinsonism, dystonia, myoclonus, postural tremor)

                        Seizures

                        Dementia

            Pes Cavus

            Osteoporosis

            Premature atherosclerosis

 

DIAGNOSIS

 

The diagnosis of CTX is mainly based on clinical features, biochemical testing, neuroimaging, and molecular genetic analysis. Since clinical presentation can be variable in type, severity, and timing, diagnosis is often delayed. Patients with CTX include having high plasma cholestanol concentration (five- to ten-fold greater than normal), normal-to-low plasma cholesterol concentration, elevated urine bile alcohol concentration, elevated plasma bile alcohol concentration, decreased CDCA level, and increased levels of cholestanol and apolipoprotein B in cerebrospinal fluid (10,17). Cholesterol concentration in tissue is increased but plasma cholesterol levels are low to normal (27). Brain MRI features are strongly suggestive of diagnosis and include bilateral hyperintensity of the dentate nuclei, diffuse cerebral and cerebellar atrophy, and white matter signal abnormalities. Brain MRI spectroscopy shows decreased n-acetylaspartate and increased lactate signals, suggestive of widespread axonal damage and cerebral mitochondrial dysfunction (20). Although not necessary for initiation of treatment, molecular testing provides definitive confirmation of CTX. Various mutations in all nine exons and in introns 2,4,6,7 and 8 of the CYP27A1 gene have been described worldwide (8). Mignarri et al (22) developed a suspicion index to be used by clinicians to calculate CTX prediction score. They proposed using family history, systemic and neurologic features as diagnostic indicators, classified as very strong (score of 100; e.g., tendon xanthoma or, sibling with CTX); strong (score of 50; e.g., juvenile cataract, chronic diarrhea, prolonged neonatal cholestasis, ataxia, MRI alterations, intellectual disability and/or psychiatric disturbances); or moderate (score of 25; e.g., early osteoporosis, epilepsy, parkinsonism or polyneuropathy. A total score ≥ 100 warrants serum cholestanol assessment, and elevated cholestanol or total score ≥ 200, with one very strong or four strong indicators, warrants CYP27A1 gene analysis (22). Quantification of bile acid precursor 7α-hydroxy-4-cholesten-3-one has been proposed to be a rapid, convenient diagnostic test for CTX (28). Early detection of ketosterol bile acid precursors can play important role in early detection through newborn screening (29).

 

MANAGEMENT

 

Early initiation of oral chenodeoxy-cholic-acid (CDCA) therapy at a dose of 250 mg given 3 times daily for adults and 15 mg/kg/d for children is the treatment of choice for neurological and non-neurological symptoms such as diarrhea (30-32), however it still does not have U.S. Food and Drug Administration approval for CTX. FDA has granted it an orphan drug designation for use in CTX.  CDCA has been approved in the European Union to treat adults and children with CTX over 1 month of age. This treatment suppresses synthesis of cholesterol, cholestanol, bile alcohols, and bile acids and alleviates clinical symptoms if started at an early age (33,34); however it has not been shown to improve bone mineral density in affected patients (35). The treatment is most effective when started early (31), and patients diagnosed later in life with significant neurological disease may progress despite CDCA therapy. A retrospective cohort study in 79 Dutch patients with CTX showed that the MRI brain remained normal even after 25 year of follow-up treatment if therapy was started in young patients with a normal MRI at diagnosis. Treatment with cholic acid has also been shown to be effective, especially in patients with side effects to CDCA therapy (36,37). HMG-CoA-reductase-inhibitors along with CDCA can improve lipoprotein metabolism, inhibit cholesterol synthesis, and reduce plasma levels of cholestanol (38). Surgical excision of bilateral tendon may worsen the gait imbalance and cannot prevent the deterioration of neurologically affected patients (39). Lumbreras et al (40) showed that adeno-associated virus (AAV) vectors expressing CYP27A1 restored bile acid metabolism and normalized the concentration of most bile acids in plasma in a CTX mouse model, making gene therapy a feasible option.

 

SCREENING

 

Screening of all first-degree relatives, when feasible, should be considered as early detection and treatment are the most important aspects for preventing morbidity and mortality (41). A recent pilot study has also shown promising option of screening for CTX as part of newborn screening (42). A recent study on de-identified dried blood spots of 20,076 newborns from the 2019 cohort of the biobank of the Dutch newborn screening program (Dutch National Institute for Puublic Health and the Environment, Bilthoven, The Netherlands) showed that metabolite ratios bile alcohol cholestanetetrol glucuronide (GlcA-tetrol)/ tauro-chenodeoxycholic acid (t-CDCA) to be informative biomarker, paving way for introduction of CTX newborn screen (43).

 

CONCLUSIONS

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. Clinical manifestations include neonatal cholestatic jaundice, infantile-onset diarrhea with failure to thrive, childhood-onset cataracts, tendon xanthomas, and progressive neurological dysfunction including intellectual disability, dementia, and psychiatric symptoms. Treatment with CDCA is treatment of choice, however cholic acid, HMG-Co-A reductase inhibitors, and surgical excision also play a role in management. Timely detection and treatment are the key to prevent severe morbidity and mortality in these patients.

 

REFERENCES

 

  1. Salen G, Steiner RD. Epidemiology, diagnosis, and treatment of cerebrotendinous xanthomatosis (CTX). J Inherit Metab Dis 2017; 40:771-781
  2. Chen C, Zhang Y, Wu H, Sun YM, Cai YH, Wu JJ, Wang J, Gong LY, Ding ZT. Clinical and molecular genetic features of cerebrotendinous xanthomatosis patients in Chinese families. Metab Brain Dis 2017; 32:1609-1618
  3. Gallus GN, Dotti MT, Federico A. Clinical and molecular diagnosis of cerebrotendinous xanthomatosis with a review of the mutations in the CYP27A1 gene. Neurol Sci 2006; 27:143-149
  4. Verrips A, Hoefsloot LH, Steenbergen GC, Theelen JP, Wevers RA, Gabreels FJ, van Engelen BG, van den Heuvel LP. Clinical and molecular genetic characteristics of patients with cerebrotendinous xanthomatosis. Brain2000; 123 ( Pt 5):908-919
  5. van Bogaert L, Scherer, H.J. and Epstein, E. . Une forme cérébrale de la cholestérinose généralisée. Paris: Masson et Cie.
  6. Swanson PD. Cerebrotendinous xanthomatosis. N Engl J Med 1968; 278:857
  7. Lorincz MT, Rainier S, Thomas D, Fink JK. Cerebrotendinous xanthomatosis: possible higher prevalence than previously recognized. Arch Neurol 2005; 62:1459-1463
  8. Pramparo T, Steiner RD, Rodems S, Jenkinson C. Allelic prevalence and geographic distribution of cerebrotendinous xanthomatosis. Orphanet J Rare Dis 2023; 18:13
  9. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 2003; 72:137-174
  10. Lorbek G, Lewinska M, Rozman D. Cytochrome P450s in the synthesis of cholesterol and bile acids--from mouse models to human diseases. FEBS J 2012; 279:1516-1533
  11. Bjorkhem I, Hansson M. Cerebrotendinous xanthomatosis: an inborn error in bile acid synthesis with defined mutations but still a challenge. Biochem Biophys Res Commun 2010; 396:46-49
  12. Salen G, Berginer V, Shore V, Horak I, Horak E, Tint GS, Shefer S. Increased concentrations of cholestanol and apolipoprotein B in the cerebrospinal fluid of patients with cerebrotendinous xanthomatosis. Effect of chenodeoxycholic acid. N Engl J Med 1987; 316:1233-1238
  13. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 1991; 266:7779-7783
  14. Cruysberg JR. Cerebrotendinous xanthomatosis: juvenile cataract and chronic diarrhea before the onset of neurologic disease. Arch Neurol 2002; 59:1975
  15. van Heijst AF, Verrips A, Wevers RA, Cruysberg JR, Renier WO, Tolboom JJ. Treatment and follow-up of children with cerebrotendinous xanthomatosis. Eur J Pediatr 1998; 157:313-316
  16. Clayton PT, Verrips A, Sistermans E, Mann A, Mieli-Vergani G, Wevers R. Mutations in the sterol 27-hydroxylase gene (CYP27A) cause hepatitis of infancy as well as cerebrotendinous xanthomatosis. J Inherit Metab Dis 2002; 25:501-513
  17. Moghadasian MH. Cerebrotendinous xanthomatosis: clinical course, genotypes and metabolic backgrounds. Clin Invest Med 2004; 27:42-50
  18. Keren Z, Falik-Zaccai TC. Cerebrotendinous xanthomatosis (CTX): a treatable lipid storage disease. Pediatr Endocrinol Rev 2009; 7:6-11
  19. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D. Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis. Metabolism 1993; 42:69-74
  20. Berginer VM, Abeliovich D. Genetics of cerebrotendinous xanthomatosis (CTX): an autosomal recessive trait with high gene frequency in Sephardim of Moroccan origin. Am J Med Genet 1981; 10:151-157
  21. Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis: a comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J Rare Dis 2014; 9:179
  22. Mignarri A, Gallus GN, Dotti MT, Federico A. A suspicion index for early diagnosis and treatment of cerebrotendinous xanthomatosis. J Inherit Metab Dis 2014; 37:421-429
  23. Stelten BML, van de Warrenburg BPC, Wevers RA, Verrips A. Movement disorders in cerebrotendinous xanthomatosis. Parkinsonism Relat Disord 2019; 58:12-16
  24. Bjorkhem I, Andersson O, Diczfalusy U, Sevastik B, Xiu RJ, Duan C, Lund E. Atherosclerosis and sterol 27-hydroxylase: evidence for a role of this enzyme in elimination of cholesterol from human macrophages. Proc Natl Acad Sci U S A 1994; 91:8592-8596
  25. Valdivielso P, Calandra S, Duran JC, Garuti R, Herrera E, Gonzalez P. Coronary heart disease in a patient with cerebrotendinous xanthomatosis. J Intern Med 2004; 255:680-683
  26. Fujiyama J, Kuriyama M, Arima S, Shibata Y, Nagata K, Takenaga S, Tanaka H, Osame M. Atherogenic risk factors in cerebrotendinous xanthomatosis. Clin Chim Acta 1991; 200:1-11
  27. Salen G. Cholestanol deposition in cerebrotendinous xanthomatosis. A possible mechanism. Ann Intern Med1971; 75:843-851
  28. DeBarber AE, Connor WE, Pappu AS, Merkens LS, Steiner RD. ESI-MS/MS quantification of 7alpha-hydroxy-4-cholesten-3-one facilitates rapid, convenient diagnostic testing for cerebrotendinous xanthomatosis. Clin Chim Acta 2010; 411:43-48
  29. DeBarber AE, Luo J, Star-Weinstock M, Purkayastha S, Geraghty MT, Chiang JP, Merkens LS, Pappu AS, Steiner RD. A blood test for cerebrotendinous xanthomatosis with potential for disease detection in newborns. J Lipid Res 2014; 55:146-154
  30. Sekijima Y, Koyama S, Yoshinaga T, Koinuma M, Inaba Y. Nationwide survey on cerebrotendinous xanthomatosis in Japan. J Hum Genet 2018; 63:271-280
  31. Duell PB, Salen G, Eichler FS, DeBarber AE, Connor SL, Casaday L, Jayadev S, Kisanuki Y, Lekprasert P, Malloy MJ, Ramdhani RA, Ziajka PE, Quinn JF, Su KG, Geller AS, Diffenderfer MR, Schaefer EJ. Diagnosis, treatment, and clinical outcomes in 43 cases with cerebrotendinous xanthomatosis. Journal of clinical lipidology2018; 12:1169-1178
  32. Stelten BML, Huidekoper HH, van de Warrenburg BPC, Brilstra EH, Hollak CEM, Haak HR, Kluijtmans LAJ, Wevers RA, Verrips A. Long-term treatment effect in cerebrotendinous xanthomatosis depends on age at treatment start. Neurology 2019; 92:e83-e95
  33. Salen G, Meriwether TW, Nicolau G. Chenodeoxycholic acid inhibits increased cholesterol and cholestanol synthesis in patients with cerebrotendinous xanthomatosis. Biochem Med 1975; 14:57-74
  34. Berginer VM, Salen G, Shefer S. Long-term treatment of cerebrotendinous xanthomatosis with chenodeoxycholic acid. N Engl J Med 1984; 311:1649-1652
  35. Zubarioglu T, Bilen IP, Kiykim E, Dogan BB, Enver EO, Cansever MS, Zeybek ACA. Evaluation of the effect of chenodeoxycholic acid treatment on skeletal system findings in patients with cerebrotendinous xanthomatosis. Turk Pediatri Ars 2019; 54:113-118
  36. Koopman BJ, Wolthers BG, van der Molen JC, Waterreus RJ. Bile acid therapies applied to patients suffering from cerebrotendinous xanthomatosis. Clin Chim Acta 1985; 152:115-122
  37. Mandia D, Chaussenot A, Besson G, Lamari F, Castelnovo G, Curot J, Duval F, Giral P, Lecerf JM, Roland D, Pierdet H, Douillard C, Nadjar Y. Cholic acid as a treatment for cerebrotendinous xanthomatosis in adults. J Neurol 2019; 266:2043-2050
  38. Kuriyama M, Tokimura Y, Fujiyama J, Utatsu Y, Osame M. Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. J Neurol Sci 1994; 125:22-28
  39. Moghadasian MH, Salen G, Frohlich JJ, Scudamore CH. Cerebrotendinous xanthomatosis: a rare disease with diverse manifestations. Arch Neurol 2002; 59:527-529
  40. Lumbreras S, Ricobaraza A, Baila-Rueda L, Gonzalez-Aparicio M, Mora-Jimenez L, Uriarte I, Bunuales M, Avila MA, Monte MJ, Marin JJG, Cenarro A, Gonzalez-Aseguinolaza G, Hernandez-Alcoceba R. Gene supplementation of CYP27A1 in the liver restores bile acid metabolism in a mouse model of cerebrotendinous xanthomatosis. Mol Ther Methods Clin Dev 2021; 22:210-221
  41. Wilson DP, Patni N. Should children with chronic diarrhea be referred to a lipid clinic? Journal of clinical lipidology 2018; 12:1099-1101
  42. DeBarber AE, Kalfon L, Fedida A, Fleisher Sheffer V, Ben Haroush S, Chasnyk N, Shuster Biton E, Mandel H, Jeffries K, Shinwell ES, Falik-Zaccai TC. Newborn screening for cerebrotendinous xanthomatosis is the solution for early identification and treatment. J Lipid Res 2018; 59:2214-2222
  43. Vaz FM, Jamal Y, Barto R, Gelb MH, DeBarber AE, Wevers RA, Nelen MR, Verrips A, Bootsma AH, Bouva MJ, Kleise N, van der Zee W, He T, Salomons GS, Huidekoper HH. Newborn screening for Cerebrotendinous Xanthomatosis: A retrospective biomarker study using both flow-injection and UPLC-MS/MS analysis in 20,000 newborns. Clin Chim Acta 2023; 539:170-174

 

Cerebrotendinous Xanthomatosis

ABSTRACT

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder caused by homozygous or compound heterozygous mutations in the CYP27A1 gene. These patients lack mitochondrial sterol 27-hydroxylase enzyme, which is responsible for conversion of cholesterol to cholic acid and chenodeoxycholic acid (CDCA) in bile acid synthesis pathway. CYP27A1 mutation leads to decreased synthesis of bile acid, excess production of cholestanol, and consequent accumulation of cholestanol in tissues, including brain, leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts. The clinical manifestations usually start at infancy and develop during the first and second decades of life. The diagnosis of CTX is based on clinical findings, biochemical testing, and neuroimaging. Molecular genetic analysis although not necessary for initiation of treatment, provides definitive confirmation of CTX. Early initiation of CDCA is the treatment of choice for neurological and non-neurological symptoms of CTX and treatment with cholic acid has also been shown to be effective for non-neurological symptoms.

 

INTRODUCTION

 

Cerebrotendinous xanthomatosis (CTX; OMIM#213700) is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. It is caused by homozygous or compound heterozygous mutation in the CYP27A1 gene located on chromosome 2q33-qter. CYP27A1 gene encodes sterol 27-hydroxylase which is involved in bile acid synthesis. Over 100 different mutations (missense, deletions, insertions, splice site, and nonsense mutations) of the CYP27A1 gene have been reported worldwide in patients of different ethnic origin (1,2). About 50% of these mutations were found in the region of exons 6–8 of the CYP27A1 gene, however here is no genotype phenotype correlation (3,4). The metabolic pathway for bile acids synthesis is shown in the figure 1.

 

Figure 1. Bile acids are synthesized from cholesterol in the liver through two pathways: the classic pathway and the alternative pathway. The bile acid synthesis mainly produces two primary bile acids, cholic acid and chenodeoxycholic acid (CDCA). In the classic pathway, the mitochondrial sterol 27-hydroxylase (CYP27A1) catalyzes the steroid side-chain oxidation in both CA and CDCA synthesis. In the alternative pathway, CYP27A1 catalyzes the first step to convert cholesterol to 27-hydroxycholesterol which eventually is converted to CDCA.

 

CTX was first described by Van Bogart, Scherer and Epstein in 1937 as a condition characterized by early development of cataracts and large tendon xanthomas and later by progressive neurologic impairment (5,6). The incidence of CTX is estimated to be 3 to 5 per 100,000 people (1,7). The prevalence is estimated to be highest in Asians (1:44,407-93,084), intermediate in Europeans, Americans, and Africans/African Americans (1:70,795-233,597) and lowest in the Finnish population (1:3,388,767) (8). 

 

ETIOLOGY AND PATHOGENESIS

 

Patients with CTX lack mitochondrial sterol 27-hydroxylase, which is an important enzyme in both the alternative and classic bile acid synthesis pathways (9). The biochemical defect prevents the synthesis of chenodeoxycholic acid (CDCA) and cholic acid (10).The absence of the negative feedback mechanism of CDCA on 7a-hydroxylase, the rate limiting step, leads to the accumulation of cholestanol and its precursor 7α-hydroxy-4-cholesten-3-one (Figure 1) (9-11). Bile acid alcohols are glucoronidated and can be found in significantly increased amounts in blood, urine, and feces of untreated CTX subjects. It has been hypothesized that the increased bile alcohol levels may lead to disruption of the blood–brain barrier (BBB), and that increased permeability of BBB may be caused by circulating bile alcohol glucuronides (12). Cholestanol and cholesterol accumulate in many tissues, including brain (primarily white matter), leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts (13).

 

CLINICAL FEATURES

 

Patients with CTX present diverse manifestations with multi-organ involvement and a broad range of neurological and non-neurological symptoms (Table 1). The clinical manifestations usually start at infancy and develop during the first and second decades of life. Neonatal cholestatic jaundice and infantile-onset diarrhea with failure to thrive may be the earliest clinical manifestation (14-16). Childhood-onset cataracts are a common early symptom, described in 92% patients with CTX. Cataracts precede neurological signs and tendon xanthoma, and if present, are considered useful for early diagnosis (14,16,17). Tendon xanthomas have been documented in 71% patients with CTX and can appear in first, second or third decade. They are common in Achilles tendon, but can also be seen in fingers, tibial tuberosity, and triceps (17,18). Pes cavus deformity has also been described (19,20). Neurological dysfunction is almost always present in patients with CTX and usually occurs in late adolescents and early adulthood. The range of symptoms are wide and include intellectual disability; dementia; psychiatric symptoms (i.e., behavioral changes, depression, agitation, hallucination, and suicide attempts); pyramidal signs (spasticity, hyperreflexia, extensor plantar responses); cerebellar signs (progressive ataxia, dysarthria); movement disorder (parkinsonism, dystonia, myoclonus, postural tremor); seizures; and peripheral neuropathy (1,4,21-23). Osteoporosis, heart involvement, and premature atherosclerosis have also been described later in life (19,24-26).

 

Table 1. Clinical Features of Cerebrotendinous Xanthomatosis

Early childhood

            Neonatal cholestatic jaundice

            Chronic diarrhea

            Cataracts

            Developmental Delay

Late childhood

            Tendon xanthomas

            Psychiatric disorders

Adulthood

            Neurological dysfunction

                        Pyramidal signs (spasticity, hyperreflexia, extensor plantar responses)

                        Cerebellar signs (ataxia, dysarthria)

                        Movement disorder (parkinsonism, dystonia, myoclonus, postural tremor)

                        Seizures

                        Dementia

            Pes Cavus

            Osteoporosis

            Premature atherosclerosis

 

DIAGNOSIS

 

The diagnosis of CTX is mainly based on clinical features, biochemical testing, neuroimaging, and molecular genetic analysis. Since clinical presentation can be variable in type, severity, and timing, diagnosis is often delayed. Patients with CTX include having high plasma cholestanol concentration (five- to ten-fold greater than normal), normal-to-low plasma cholesterol concentration, elevated urine bile alcohol concentration, elevated plasma bile alcohol concentration, decreased CDCA level, and increased levels of cholestanol and apolipoprotein B in cerebrospinal fluid (10,17). Cholesterol concentration in tissue is increased but plasma cholesterol levels are low to normal (27). Brain MRI features are strongly suggestive of diagnosis and include bilateral hyperintensity of the dentate nuclei, diffuse cerebral and cerebellar atrophy, and white matter signal abnormalities. Brain MRI spectroscopy shows decreased n-acetylaspartate and increased lactate signals, suggestive of widespread axonal damage and cerebral mitochondrial dysfunction (20). Although not necessary for initiation of treatment, molecular testing provides definitive confirmation of CTX. Various mutations in all nine exons and in introns 2,4,6,7 and 8 of the CYP27A1 gene have been described worldwide (8). Mignarri et al (22) developed a suspicion index to be used by clinicians to calculate CTX prediction score. They proposed using family history, systemic and neurologic features as diagnostic indicators, classified as very strong (score of 100; e.g., tendon xanthoma or, sibling with CTX); strong (score of 50; e.g., juvenile cataract, chronic diarrhea, prolonged neonatal cholestasis, ataxia, MRI alterations, intellectual disability and/or psychiatric disturbances); or moderate (score of 25; e.g., early osteoporosis, epilepsy, parkinsonism or polyneuropathy. A total score ≥ 100 warrants serum cholestanol assessment, and elevated cholestanol or total score ≥ 200, with one very strong or four strong indicators, warrants CYP27A1 gene analysis (22). Quantification of bile acid precursor 7α-hydroxy-4-cholesten-3-one has been proposed to be a rapid, convenient diagnostic test for CTX (28). Early detection of ketosterol bile acid precursors can play important role in early detection through newborn screening (29).

 

MANAGEMENT

 

Early initiation of oral chenodeoxy-cholic-acid (CDCA) therapy at a dose of 250 mg given 3 times daily for adults and 15 mg/kg/d for children is the treatment of choice for neurological and non-neurological symptoms such as diarrhea (30-32), however it still does not have U.S. Food and Drug Administration approval for CTX. FDA has granted it an orphan drug designation for use in CTX.  CDCA has been approved in the European Union to treat adults and children with CTX over 1 month of age. This treatment suppresses synthesis of cholesterol, cholestanol, bile alcohols, and bile acids and alleviates clinical symptoms if started at an early age (33,34); however it has not been shown to improve bone mineral density in affected patients (35). The treatment is most effective when started early (31), and patients diagnosed later in life with significant neurological disease may progress despite CDCA therapy. A retrospective cohort study in 79 Dutch patients with CTX showed that the MRI brain remained normal even after 25 year of follow-up treatment if therapy was started in young patients with a normal MRI at diagnosis. Treatment with cholic acid has also been shown to be effective, especially in patients with side effects to CDCA therapy (36,37). HMG-CoA-reductase-inhibitors along with CDCA can improve lipoprotein metabolism, inhibit cholesterol synthesis, and reduce plasma levels of cholestanol (38). Surgical excision of bilateral tendon may worsen the gait imbalance and cannot prevent the deterioration of neurologically affected patients (39). Lumbreras et al (40) showed that adeno-associated virus (AAV) vectors expressing CYP27A1 restored bile acid metabolism and normalized the concentration of most bile acids in plasma in a CTX mouse model, making gene therapy a feasible option.

 

SCREENING

 

Screening of all first-degree relatives, when feasible, should be considered as early detection and treatment are the most important aspects for preventing morbidity and mortality (41). A recent pilot study has also shown promising option of screening for CTX as part of newborn screening (42). A recent study on de-identified dried blood spots of 20,076 newborns from the 2019 cohort of the biobank of the Dutch newborn screening program (Dutch National Institute for Puublic Health and the Environment, Bilthoven, The Netherlands) showed that metabolite ratios bile alcohol cholestanetetrol glucuronide (GlcA-tetrol)/ tauro-chenodeoxycholic acid (t-CDCA) to be informative biomarker, paving way for introduction of CTX newborn screen (43).

 

CONCLUSIONS

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. Clinical manifestations include neonatal cholestatic jaundice, infantile-onset diarrhea with failure to thrive, childhood-onset cataracts, tendon xanthomas, and progressive neurological dysfunction including intellectual disability, dementia, and psychiatric symptoms. Treatment with CDCA is treatment of choice, however cholic acid, HMG-Co-A reductase inhibitors, and surgical excision also play a role in management. Timely detection and treatment are the key to prevent severe morbidity and mortality in these patients.

 

REFERENCES

 

  1. Salen G, Steiner RD. Epidemiology, diagnosis, and treatment of cerebrotendinous xanthomatosis (CTX). J Inherit Metab Dis 2017; 40:771-781
  2. Chen C, Zhang Y, Wu H, Sun YM, Cai YH, Wu JJ, Wang J, Gong LY, Ding ZT. Clinical and molecular genetic features of cerebrotendinous xanthomatosis patients in Chinese families. Metab Brain Dis 2017; 32:1609-1618
  3. Gallus GN, Dotti MT, Federico A. Clinical and molecular diagnosis of cerebrotendinous xanthomatosis with a review of the mutations in the CYP27A1 gene. Neurol Sci 2006; 27:143-149
  4. Verrips A, Hoefsloot LH, Steenbergen GC, Theelen JP, Wevers RA, Gabreels FJ, van Engelen BG, van den Heuvel LP. Clinical and molecular genetic characteristics of patients with cerebrotendinous xanthomatosis. Brain2000; 123 ( Pt 5):908-919
  5. van Bogaert L, Scherer, H.J. and Epstein, E. . Une forme cérébrale de la cholestérinose généralisée. Paris: Masson et Cie.
  6. Swanson PD. Cerebrotendinous xanthomatosis. N Engl J Med 1968; 278:857
  7. Lorincz MT, Rainier S, Thomas D, Fink JK. Cerebrotendinous xanthomatosis: possible higher prevalence than previously recognized. Arch Neurol 2005; 62:1459-1463
  8. Pramparo T, Steiner RD, Rodems S, Jenkinson C. Allelic prevalence and geographic distribution of cerebrotendinous xanthomatosis. Orphanet J Rare Dis 2023; 18:13
  9. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 2003; 72:137-174
  10. Lorbek G, Lewinska M, Rozman D. Cytochrome P450s in the synthesis of cholesterol and bile acids--from mouse models to human diseases. FEBS J 2012; 279:1516-1533
  11. Bjorkhem I, Hansson M. Cerebrotendinous xanthomatosis: an inborn error in bile acid synthesis with defined mutations but still a challenge. Biochem Biophys Res Commun 2010; 396:46-49
  12. Salen G, Berginer V, Shore V, Horak I, Horak E, Tint GS, Shefer S. Increased concentrations of cholestanol and apolipoprotein B in the cerebrospinal fluid of patients with cerebrotendinous xanthomatosis. Effect of chenodeoxycholic acid. N Engl J Med 1987; 316:1233-1238
  13. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 1991; 266:7779-7783
  14. Cruysberg JR. Cerebrotendinous xanthomatosis: juvenile cataract and chronic diarrhea before the onset of neurologic disease. Arch Neurol 2002; 59:1975
  15. van Heijst AF, Verrips A, Wevers RA, Cruysberg JR, Renier WO, Tolboom JJ. Treatment and follow-up of children with cerebrotendinous xanthomatosis. Eur J Pediatr 1998; 157:313-316
  16. Clayton PT, Verrips A, Sistermans E, Mann A, Mieli-Vergani G, Wevers R. Mutations in the sterol 27-hydroxylase gene (CYP27A) cause hepatitis of infancy as well as cerebrotendinous xanthomatosis. J Inherit Metab Dis 2002; 25:501-513
  17. Moghadasian MH. Cerebrotendinous xanthomatosis: clinical course, genotypes and metabolic backgrounds. Clin Invest Med 2004; 27:42-50
  18. Keren Z, Falik-Zaccai TC. Cerebrotendinous xanthomatosis (CTX): a treatable lipid storage disease. Pediatr Endocrinol Rev 2009; 7:6-11
  19. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D. Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis. Metabolism 1993; 42:69-74
  20. Berginer VM, Abeliovich D. Genetics of cerebrotendinous xanthomatosis (CTX): an autosomal recessive trait with high gene frequency in Sephardim of Moroccan origin. Am J Med Genet 1981; 10:151-157
  21. Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis: a comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J Rare Dis 2014; 9:179
  22. Mignarri A, Gallus GN, Dotti MT, Federico A. A suspicion index for early diagnosis and treatment of cerebrotendinous xanthomatosis. J Inherit Metab Dis 2014; 37:421-429
  23. Stelten BML, van de Warrenburg BPC, Wevers RA, Verrips A. Movement disorders in cerebrotendinous xanthomatosis. Parkinsonism Relat Disord 2019; 58:12-16
  24. Bjorkhem I, Andersson O, Diczfalusy U, Sevastik B, Xiu RJ, Duan C, Lund E. Atherosclerosis and sterol 27-hydroxylase: evidence for a role of this enzyme in elimination of cholesterol from human macrophages. Proc Natl Acad Sci U S A 1994; 91:8592-8596
  25. Valdivielso P, Calandra S, Duran JC, Garuti R, Herrera E, Gonzalez P. Coronary heart disease in a patient with cerebrotendinous xanthomatosis. J Intern Med 2004; 255:680-683
  26. Fujiyama J, Kuriyama M, Arima S, Shibata Y, Nagata K, Takenaga S, Tanaka H, Osame M. Atherogenic risk factors in cerebrotendinous xanthomatosis. Clin Chim Acta 1991; 200:1-11
  27. Salen G. Cholestanol deposition in cerebrotendinous xanthomatosis. A possible mechanism. Ann Intern Med1971; 75:843-851
  28. DeBarber AE, Connor WE, Pappu AS, Merkens LS, Steiner RD. ESI-MS/MS quantification of 7alpha-hydroxy-4-cholesten-3-one facilitates rapid, convenient diagnostic testing for cerebrotendinous xanthomatosis. Clin Chim Acta 2010; 411:43-48
  29. DeBarber AE, Luo J, Star-Weinstock M, Purkayastha S, Geraghty MT, Chiang JP, Merkens LS, Pappu AS, Steiner RD. A blood test for cerebrotendinous xanthomatosis with potential for disease detection in newborns. J Lipid Res 2014; 55:146-154
  30. Sekijima Y, Koyama S, Yoshinaga T, Koinuma M, Inaba Y. Nationwide survey on cerebrotendinous xanthomatosis in Japan. J Hum Genet 2018; 63:271-280
  31. Duell PB, Salen G, Eichler FS, DeBarber AE, Connor SL, Casaday L, Jayadev S, Kisanuki Y, Lekprasert P, Malloy MJ, Ramdhani RA, Ziajka PE, Quinn JF, Su KG, Geller AS, Diffenderfer MR, Schaefer EJ. Diagnosis, treatment, and clinical outcomes in 43 cases with cerebrotendinous xanthomatosis. Journal of clinical lipidology2018; 12:1169-1178
  32. Stelten BML, Huidekoper HH, van de Warrenburg BPC, Brilstra EH, Hollak CEM, Haak HR, Kluijtmans LAJ, Wevers RA, Verrips A. Long-term treatment effect in cerebrotendinous xanthomatosis depends on age at treatment start. Neurology 2019; 92:e83-e95
  33. Salen G, Meriwether TW, Nicolau G. Chenodeoxycholic acid inhibits increased cholesterol and cholestanol synthesis in patients with cerebrotendinous xanthomatosis. Biochem Med 1975; 14:57-74
  34. Berginer VM, Salen G, Shefer S. Long-term treatment of cerebrotendinous xanthomatosis with chenodeoxycholic acid. N Engl J Med 1984; 311:1649-1652
  35. Zubarioglu T, Bilen IP, Kiykim E, Dogan BB, Enver EO, Cansever MS, Zeybek ACA. Evaluation of the effect of chenodeoxycholic acid treatment on skeletal system findings in patients with cerebrotendinous xanthomatosis. Turk Pediatri Ars 2019; 54:113-118
  36. Koopman BJ, Wolthers BG, van der Molen JC, Waterreus RJ. Bile acid therapies applied to patients suffering from cerebrotendinous xanthomatosis. Clin Chim Acta 1985; 152:115-122
  37. Mandia D, Chaussenot A, Besson G, Lamari F, Castelnovo G, Curot J, Duval F, Giral P, Lecerf JM, Roland D, Pierdet H, Douillard C, Nadjar Y. Cholic acid as a treatment for cerebrotendinous xanthomatosis in adults. J Neurol 2019; 266:2043-2050
  38. Kuriyama M, Tokimura Y, Fujiyama J, Utatsu Y, Osame M. Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. J Neurol Sci 1994; 125:22-28
  39. Moghadasian MH, Salen G, Frohlich JJ, Scudamore CH. Cerebrotendinous xanthomatosis: a rare disease with diverse manifestations. Arch Neurol 2002; 59:527-529
  40. Lumbreras S, Ricobaraza A, Baila-Rueda L, Gonzalez-Aparicio M, Mora-Jimenez L, Uriarte I, Bunuales M, Avila MA, Monte MJ, Marin JJG, Cenarro A, Gonzalez-Aseguinolaza G, Hernandez-Alcoceba R. Gene supplementation of CYP27A1 in the liver restores bile acid metabolism in a mouse model of cerebrotendinous xanthomatosis. Mol Ther Methods Clin Dev 2021; 22:210-221
  41. Wilson DP, Patni N. Should children with chronic diarrhea be referred to a lipid clinic? Journal of clinical lipidology 2018; 12:1099-1101
  42. DeBarber AE, Kalfon L, Fedida A, Fleisher Sheffer V, Ben Haroush S, Chasnyk N, Shuster Biton E, Mandel H, Jeffries K, Shinwell ES, Falik-Zaccai TC. Newborn screening for cerebrotendinous xanthomatosis is the solution for early identification and treatment. J Lipid Res 2018; 59:2214-2222
  43. Vaz FM, Jamal Y, Barto R, Gelb MH, DeBarber AE, Wevers RA, Nelen MR, Verrips A, Bootsma AH, Bouva MJ, Kleise N, van der Zee W, He T, Salomons GS, Huidekoper HH. Newborn screening for Cerebrotendinous Xanthomatosis: A retrospective biomarker study using both flow-injection and UPLC-MS/MS analysis in 20,000 newborns. Clin Chim Acta 2023; 539:170-174

 

Defects of Thyroid Hormone Transport in Serum

ABSTRACT

 

Inherited abnormalities of thyroid hormone-binding proteins are not uncommon and can predominate in some ethnic groups. They alter the number of iodothyronines present in serum and, although the concentration of free hormones remains unaltered, routine measurement can give erroneous results. With a single exception, inherited defects in thyroxine-binding globulin (TBG), are X-chromosome linked and thus, the full phenotype is expressed mostly in males. Partial TBG deficiency is more common than complete deficiency. High frequency of variants TBGs have been identified in African Blacks, Australian Aborigine, and Eskimos. Most defects producing TBG deficiency are caused by mutations in the structural gene. However, inherited X-linked partial deficiency can occur as the consequence of mutations of a gene enhancer. Inherited forms of TBG excess are all caused by gene duplication or triplication. Mutations in the transthyretin (TTR) gene producing a molecule with increased affinity for T4 are relatively rare. A variant TTR produces transient hyperthyroxinemia during non-thyroidal illness.     Mutations of the human serum albumin (HSA) gene produce increased concentration of serum T4, a condition known as familial dysalbuminemic hyperthyroxinemia (FDH). They are relatively more common in individuals of Hispanic origin. They cause an increase in serum T4 owing to increased affinity for this iodothyronine but high concentrations in free T4 observed in direct measurement by some commercial methods are erroneous. A variant with increased affinity for T3 has been also identified.

 

INTRODUCTION

 

Abnormalities in the serum proteins that transport thyroid hormone do not alter the metabolic state and do not cause thyroid disease.  However, they do produce alterations in thyroid hormone concentration in serum and when unrecognized have led to inappropriate treatment.  When the abnormality is the consequence of altered synthesis, secretion or stability of the variant serum protein, the free thyroid hormone level estimated by most of the clinically available techniques remains within the range of normal. In contrast, when the defect results in a significant alteration of the affinity of the variant protein for the hormone, estimates of the free thyroid hormone level often give erroneous results and thus, it is prudent to measure the free hormone concentration by more direct methods such as equilibrium dialysis or ultrafiltration.  This is also true in cases of complete TBG deficiency, in whom the estimation of free thyroid hormone level in serum by indirect methods, or using iodothyronine analogs as tracers, can also give erroneous results.

 

The existence of inherited defects of serum transport of thyroid hormone was first recognized in 1959 with the report of TBG-excess by Beierwaltes and Robbins (1).  Genetic variants for each of the three major thyroid hormone transport proteins have since been described and in subsequent years, the molecular basis of a number of these defects has been identified (2).  Clinically, these defects usually manifest as either euthyroid hyperthyroxinemia or hypothyroxinemia and more rarely, isolated hypertriiodothyroninemia (3).  Associated abnormalities such as thyrotoxicosis, hypothyroidism, goiter, and familial hyperlipidemia are usually coincidental (4).  However, individuals with thyroid disorders are more likely to undergo thyroid testing leading to the fortuitous detection of a thyroid hormone transport defect.

 

THYROXINE-BINDING GLOBULIN (TBG) DEFECTS

 

Familial TBG abnormalities are inherited as X-chromosome linked traits (5, 6), compatible with the location of the TBG (SERPINA7) gene on the long arm of the X-chromosome (Xq22.2) (7, 8).  This mode of inheritance also suggests that the defects involve the TBG gene proper, rather than the rate of TBG disposal, as long ago postulated (5). The normal, common type TBG (TBG-N or TBG-C), has a high affinity for iodothyronines [affinity constants (Ka): 10-10 M-1 for T4 and 10-9 M-1 for T3] and binds 75% of the total T4 and T3 circulating in blood. Thus, with a single exception [HSA R218P and R218S (9-11), see below], among the inherited abnormalities of thyroid hormone transport proteins, those involving the TBG molecule produce usually more profound alterations of thyroid hormone concentration in serum.

 

Clinically TBG defects are classified according to the level of TBG in serum of affected hemizygotes (XY males or XO females, that express only the mutant allele): complete TBG deficiency (TBG-CD), partial TBG deficiency (TBG-PD) and TBG excess (TBG-E).  In families with TBG-CD, affected males have no detectable TBG and carrier females (mothers or daughters) have on the average half the normal TBG concentration (4).  In families with partially TBG deficient males, the mean TBG concentration in heterozygous females is usually above half the normal.  Serum TBG concentration in males with TBG-E is 2 to 4-fold the normal mean and that in the corresponding carrier females, is slightly higher than half that of the affected males.  These observations indicate an equal contribution of cells expressing the normal and mutant TBG genes. On rare occasions, selective inactivation of the X-chromosome has been the cause of manifestations of the complete defect (hemizygous phenotype) in heterozygous females (12, 13).

 

Inherited TBG defects can be further characterized by the level of denatured TBG (dnTBG) in serum and the physicochemical properties of the molecule.  The latter can be easily determined without the need of purification. These properties are: (a) immunologic identity; (b) isoelectric focusing (IEF) pattern; (c) rate of inactivation when exposed to various temperatures and pH; and (d) affinity for the ligands, T4 and T3.  More precise identification of TBG defects requires sequencing of the TBG gene.

 

MiP a Subject With TBG-CD

 

The proposita, a phenotypic female, was 13 years old when first seen because of retarded growth, amenorrhea and absence of secondary sexual traits.  She was the first sibling of a second marriage for both parents. The family included a younger brother and four older half-siblings, two maternal and two paternal. The proposita was born to her 30-year-old mother after full-term, uncomplicated pregnancy. Infancy and early childhood development were normal until 4 years of age when it became apparent that she was shorter than her peers. She was 12 years of age when a low protein bound iodine (PBI, then a measure of T4) of 2.2 µg/dl (normal range 4.0-8.0) was noted and treatment with 120 mg of desiccated thyroid (equivalent to 200µg L-T4) daily was initiated. Since, during the ensuing 6 months, no change in her growth rate occurred and because PBI remained unchanged (2.0 µg/dl), the dose of desiccated thyroid was increased to 180 mg/day. This produced restlessness, perturbed sleep and deterioration of school performance necessitating discontinuation of thyroid hormone treatment. No family history of thyroid disease or short stature was elicited and the parents’ denied consanguinity.

 

On physical examination, the patient appeared younger than her chronological age, was short (137 cm) and showed no signs of sexual development.  She had a webbed neck, low nuchal hairline, bilateral eyelid ptosis, shield-shaped chest, increased carrying angle and short 4th metacarpals and metatarsals. The thyroid gland was normal in size and consistency.

 

Buccal smear was negative for Barr bodies and karyotyping revealed 45 chromosomes consistent with XO Turner's syndrome. No chromosomal abnormalities were found in lymphocytes from the mother and father. Bone age was 12 years and X-ray of the scull showed a mild degree of hypertelorism. PBI and butanol extractable iodine were low at 2.0 and 1.8 µg/dl, respectively. Resin-T3 uptake was high at 59.9% (normal range 25-35%) indicating reduced TBG-binding capacity. A 24-hour thyroidal radioiodide uptake was normal at 29%, basal metabolic rate was +20% (normal range -10 to +20) and TG autoantibodies were not present.  Serum cortisol was normal as were the responses to ACTH and metyrapone.  Basal growth hormone concentration was normal at 8.0 ng/ml which rose to 32 ng/ml following insulin hypoglycemia.

 

Studies were carried out in all first-degree relatives and the proposita was treated cyclically with diethylstilbestrol which produced withdrawal uterine bleeding and gradual breast development.

 

Five family members, in addition to the proposita had thyroid function tests abnormalities. Two were males and three females.  The two males (maternal grandfather and maternal half-brother) and the proposita had the lowest PBI levels and undetectable T4-binding to serum TBG.  In contrast, the three females (mother, maternal aunt and maternal half-sister) had a lesser reduction of their PBI and T4-binding capacity to TBG approximately one-half the normal mean value. The two sons of the affected grandfather (maternal uncles to the proposita) had normal PBI and T4-binding to TBG. No interference with T4-binding to TBG or other serum protein abnormalities were found in affected members of the family. In vivo T4 kinetic studies revealed a rapid extrathyroidal turnover rate but normal daily secretion and degradation, compatible with their eumetabolic state.

 

INTERPRETATION

 

The incidental identification of thyroid tests abnormalities in the proposita is typical for most subjects with TBG deficiency as well as TBG excess. So is the initial unnecessary treatment; though less frequent with the routine measurement or estimation of free T4. The inherited nature of the defect is suspected by exclusion of factors known to cause acquired TBG abnormalities and should be confirmed by the presence of similar abnormalities in members of the family. The absence of male-to-male transmission and the carrier state of all female offspring of the affected males is a typical pattern of X-chromosome linked inheritance. This is further supported by the complete TBG deficiency in individuals having a single X chromosome (males and the XO female) and only partial TBG deficiency in carrier XX females.

 

Since the publication of this family in 1968 (14), the cause of the TBG defect was identified. From the mutation identified in the TBG gene of this family [TBG Harwichport (TBG-CD H)], it can be deduced that the molecule is truncated, missing 12 amino acids at the carboxyl terminus (15).

 

Fifty six TBG variants have been so far identified and in 50 the precise defect has been determined by gene analysis. Their primary structure defect, some of their physical and chemical properties and the resulting serum T4 concentrations are summarized in Table 1 and figure 1.

 

Figure 1. Properties of some TBG variants causing partial TBG deficiency (TBG-PD). The TBG variants are: -SD, San Diego; -G, Gary; -M, Montreal, -S, slow; -A, Aborigine; -Poly, polymorphic; -Cgo, Chicago; and -Q, Quebec. For detailed description, see (1) Sarne et al (39) and Bertenshaw et al (37); (2) Murata et al (34), Mori et al (43) and Kambe et al (60); (3) Takamatsu et al (44) and Janssen et al (45); (4) Takamatsu et al (55) and Waltz et al (56); (5) Murata et al (61) and Takeda et al (47); (6) Mori et al (26) and Takeda et al (57); (7) Takamatsu et al (59) and (58); (8) Takamatsu et al (44) and Bertenshaw et al (49). [Modified from Refetoff et al (62)].

 

 

Table 1.  TBG Variants and Gene Mutations

TBG NAME

Abbreviated

name

Intron

Exon

CODON*

AMINO ACID

NUCLEOTIDE

References

WT

Variant

WT

Variant

Complete Deficiency (CD)

Milano (fam A)

CDMi†

IVS 1

fs

5' DSS

unknown

gtaagt

gttaagt

(16)

Andrews

CDAN

IVS 1

fs

5' DSS

unknown

gtaagt

gcaagt

(17)

Portuguese 1 (pt A)

CDP1

1

23

S (Ser)

X (OCH)

TCA

TAA

(18)

Yonago

CDY

1

28-29fs-51

D F

X (OPA)

GA(CT)TT

GAATT

(19)

Negev (Bedouin

CDN

1

38fs-51

T (Thr)

X (OPA)

ACT

T del

(20, 21)

Nikita (fam B)

CDNi

1

50fs-51

P (Pro)

X (OPA)

CCT

T del

(16)

Taiwanese 1

CDT1†

1

52

S (Ser)

N (Asn)

AGC

AAC

(22)

Parana

CDPa†

1

61

S (Ser)

C (Cys)

TCC

TGC

a

No name

CD6

1

165fs-168

V (Val)

X (OCH)

GTT

T del

(23)

Kankakee

CDK

IVS 2

188fs-195

3' ASS

X (OPA)

agCC

ggCC

(24)

Poland

CDPL

2

201fs-206

D (Asp)

X (OCH)

GAC

G del

(25)

Portuguese 2(pt B)

CDP2

2

223

Q (Gln)

X (OCH)

CAA

TAA

(18)

No name

CD5†

2

227

L (Leu)

P (Pro)

CTA

CCA

(26)

Portuguese 3§

CDP3

2

233

N (Asn)

I (Ile)

ACC

ATC

(27)

Berlin

CDBn

IVS 3+3

fs

intronic

unknown

intronic

79nt del

(17)

Houston

CDH

IVS 3

279fs-374

3’ ASS

X (OPA)

agAT

aaAT

(28)

Buffalo

CDB

3

280

W (Trp)

X (AMB)

TGG

TAG

(29)

Taiwanese 2

CDT2

3

280

W (Trp)

X (OPA)

TGG

TGA

(22)

Lisle

CDL

IVS 4

280fs-325

5' DSS

X (OPA)

gtaaa

ggaaa

b

Jackson (fam K)

CDJa

IVS 4

280fs-325

5' DSS

X (OPA)

gtaaa

gtaag

(30)

No name

CD7

3

283fs-301

L (Leu)

X (OPA)

TGT

G del

(15)

No name

CD8†

4

329fs-374

A (Ala)

X (OPA)

GCT

G del

(15)

Japan

CDJ

4

352fs-374

L (Leu)

X (OPA)

CTT

C del

(31, 32)

Penapolis

CDPe

4

332fs-374

K (Lys)

X (OPA)

AAG

A del

a

Kyoto§

CDKo

4

370

S (Ser)

F (Phe)

TCT

TTT

(33)

Harwichport

CDH

4

381fs-396

Y (Tyr)

X (OPA)

AGG

19nt del

(14, 15, 34)

NeuIsenburg

CDNl

4

384fs-402

L (Leu)

7 aa add

CTC

TC del

(35)

Partial Deficiency (PD)

Allentown

PDAT

1

-2

H (His)

Y (Tyr)

CAC

TAC

(36)

San Diego

PDSD†

1

23

S (Ser)

T (Thr)

TCA

ACA

(37-39)

Brasilia

PDB

1

35

R (Arg)

W (Trp)

CGG

TGG

(40)

Wanne-Eickel

PDWE

1

35

R (Arg)

E (Glu)

CGG

CAG

(41)

No name

 

1

35

R (Arg)

Q (Gln)

CGG

TGG

(41)

No name

?

1

52

S (Ser)

R (Arg)

AGT

AGA

c

No name

?

1

64

A (Ala)

D (Asp)

GCC

GAC

(42)

Korea

PDKa

1

74

E (Glu)

K (Lys)

GAG

AAG

a

Gary

PDG

1

96

I (Ile)

N (Asn)

ATC

AAC

(43)

No name

 

1

112

N (Asn)

L (Lys)

AAT

AAG

c

Montréal

PDM

1

113

A (Ala)

P (Pro)

GCC

CCC

(44, 45)

Berlin2

Bn2

1

161

T (Thr)

N (Asn)

ACC

AAC

a.

Aborigine

PDA†

2

191

A (Ala)

T (Thr)

GCA

ACA

(46, 47)

Glencoe

PDGe

2

215

V (Val)

G (Gly)

GTG

GGG

(48)

Quebec

PDQ†

4

331

H (His)

Y (Tyr)

CAT

TAT

(44, 49)

Japan (Kumamoto)

PDJ

4

363

P (Pro)

L (Leu)

CCT

CTT

(50, 51)

Heidelberg

PDHg

4

368

D (Asp)

G (Gly)

GAT

GGT

(52)

No name

?

4

381

R (Arg)

G (Gly)

AGG

GGG

c

No name

?

4

382

S (Ser)

R (Arg)

AGT

CGT

c

No name

 

enhancer

 

-

-

G

A

(53)

Other Variants

Slow

S

1

171

D (Asp)

N (Asn)

GAC

AAC

(54-56)

Polymorphism

Poly

3

283

L (Leu)

F (Phe)

TTG

TTT

(26, 57)

Chicago

CH or Cgo

3

309

Y (Tyr)

F (Phe)

TAT

TTT

(58, 59)

* Codon numbering from fist amino acid of the mature protein. The 20 amino acids of the signal peptide are numbered -1 to -20, from N- to C-terminus.  The codon at the site of mutation is followed by the codon at the site of termination of translation.

† Coexistence of TBG Poly

  • complete deficiency is uncertain as the TBG assay used was unable to detect values <10% the mean normal

¶ Also a silent mutation at codon 55:  GCA -> GCG

a Personal communication

b Personal observation

c Communicated by Pia Hermanns and Joachim Pohlenz, University of Mainz, Germany

del, delete; add, addition; aa, amino acid; fs, frame shift

Pt, patent; fam, family

IVS, intervening sequence or intron; ASS, acceptor splice site; DSS, donor splice site

 

Complete Deficiency of TBG (TBG-CD)

 

TBG-CD is defined as undetectable TBG in serum of affected hemizygous subjects or a value lesser than 0.03% the normal mean; the current limits of detection using the most sensitive radioimmunoassay (RIA) being 5ng/dl (26).  The prevalence is approximately 1:15,000 newborn males.  Twenty-seven TBG variants having this phenotype have been characterized at the gene level.  These are shown in table 1 that also contains references to the original publications. Twenty two of the 27 TBG-CDs have truncated molecules. Early termination of translation of these variants is caused in 4 by a single nucleotide substitution (TBG-CDP1, TBG-CDP2, CD5, TBG-CDB and TBG-CDT2) or by a frame shift due to one nucleotide deletion (TBG-CDY, TBG-CDN, TBG-CDNi, TBG-CD6, CD-PL, TBG-CD7, TBG-CD8, and TBG-CDJ, TBG-CDPe) or deletion of 19 nucleotides (TBG-CDH).  In 7 variants mutations occurred in introns, 6 of which are close to splice sites (TBG-CDMi, TBG-CDAN, TBG-CDK, TBGBn, TBG-CDH, TBG-CDL and TBG-CDJa).  A mutation at the acceptor splice junction caused also a frame shift producing early termination of translation in TBG-CDK (24).  In contrast a nucleotide substitutions in the 5' donor splice site of intron IV (TBG-CDL and TBG-CDJa), resulted in a complete splicing of exon 3, also producing a truncated molecule (30) and personal observation.  A similar mechanism is likely responsible for CD in TBG-CDMi, though direct experimental prove was not provided (16).  Single amino acid substitution was the cause of CD in five families (TBG-CDT1, TBG-CDPa, TBG-CD5, TBG-CDP3 and TBG-CDKo).  In TBG-CD5 Leucine-227 with a proline was shown to cause aberrant post-translational processing (45).  One TBG variant (TBG-CDNI), with two nucleotides deleted close to the carboxyl terminus, the resulting frame shift predicts an extension of the molecule by the addition of 7 nonsense residues (35).  TBG-CDJ has been so far identified only in Japanese but its allele frequency in the population remains unknown (32, 57) (Table. 1).

 

Partial Deficiency of TBG (TBG-PD)

 

This is the most common form of inherited TBG deficiency having a prevalence of 1:4,000 newborn.  Identification of heterozygous females by serum TBG measurement may be difficult because levels often overlap the normal range.  In contrast to variants with complete TBG deficiency, all TBG-PDs have missense mutations. It is possible that three of the five variants with single amino acid substitutions included in the category of TBG-CD have also partial deficiency which was not identified owing to the low sensitivity of routine assays for the measurement of TBG.  Twenty-one different mutations, producing a variable degree of reduction of TBG concentration in serum, have been identified, 20 of which involve mutations in the TBG gene proper.  They are listed in table 1.  In addition, some of these variants are unstable (TBG-PDG, TBG-PDA, TBG-PDSD, TBG-PDM TBG-PDQ and TBG-PDJ) or have lower binding affinity for T4 and T3 (TBG-PDG, TBG-PDA, TBG-PDS TBG-PDSD, TBG-PDM and TBG-PDQ), impaired intracellular transport and secretion (TBG-PDJ and TBG-CDJ) and some exhibit an abnormal migration pattern on IEF electrophoresis (TBG-PDG, TBG-PDM, and TBG-PDQ) (Fig. 1).  Variants with decreased affinity for T4 and T3 have a disproportionate reduction in hormone concentration relative to the corresponding serum TBG level (Fig. 2) and estimations of the free hormone levels by any of the index methods gives erroneous results (39, 64).  One of these variants, TBG-PDA, is found with high frequency (allele frequency of 51%) in Australian Aborigines (47).

 

Figure 2. Serum T4-bound to TBG and the concentration of TBG and denatured TBG (dnTBG) in hemizygous subjects expressing the different TBG variants. Results, graphed as mean ± SD, were normalized by expressing them as % of those for the common type TBG (TBG-C). For abbreviations used in the nomenclature of the TBG variants, see legend to figure 1. [Adapted from Janssen et al (63)].

 

A unique family with TBG-PD has been described in which inheritance of the partial deficiency was autosomal dominant with transmission of the phenotype from father to son (65).  The concentration of TBG in affected males and females was about one half the normal mean value.  The TBG had normal affinity for T4. normal IEF and heat lability.  No sequence changes were found in the entire coding arias of the gene or in the promoter region.  Although the mechanism of TBG-CD in this family is unknown an abnormality in one of the factors regulating TBG gene transcription is a distinct possibility. Further studies to determine the genetic defect have been hampered by lack of subjects’ cooperation.

 

In 5% (4 or 74) families with X-chromosome lined TBG deficiency, studied in the author’s laboratory, no mutations were identified in the entire TBG gene, including all exons, introns, untranslated regions and the promoter region of the gene, covering a total of 9.2 kb. Next-generation sequencing identified a novel single nucleotide substitution 20 kb downstream of the TBG gene in all four families. In silico analysis predicted that the variant resides within a liver-specific enhancer. In vitro studies confirmed the enhancer activity of a 2.2-kb fragment of genomic DNA containing the novel variant and showed that the mutation reduces the activity of this enhancer. The affected subjects share a haplotype of 8 Mb surrounding the mutation. Three were of known Arab ethnicity and in all four families the most recent common ancestor was estimated to be 19.5 generations ago (95% confidence intervals). This is first report of an inherited endocrine disorder caused by a mutation in an enhancer region (53).

 

TBG Excess (TBG-E)

 

TBG-E has a lower prevalence than TBG deficiency, with values obtained from neonatal screening programs from 1:6,000 to 1:40,000 (66, 67).  Considering that some newborn may have non-inherited, transient TBG excess, a conservative overall estimate of inherited TBG-E would be 1:25,000 (68).  Early sequencing of the coding and promoter regions of subjects with TBG-E failed to show any defects (69).  However, in 1995, Mori et al (70) found that gene amplification was the cause of TBG-E in two families.  Gene triplication and duplication were demonstrated by gene dosage studies using HPLC measurements of the PCR -amplified product.  As expected, hemizygous affected males had approximately 3- and 2-fold the average normal serum TBG concentration, respectively.  The presence of multiple TBG gene copies in tandem was confirmed by in situ hybridization of prometaphase and interphase chromosomes from an affected male (Fig. 3).

Figure 3. Hybridization in prometaphase chromosomes of cultured skin fibroblasts obtained from an affected male with TBG-E. A complete TBG cDNA was used as a probe. Three TBG gene copies are seen in tandem with each exon clearly identified in the starched chromosome shown in panel B.

 

 

TBG Variants with Unaltered TBG Concentrations in Serum

 

Five TBG variants have been identified that present with normal or slight and clinically insignificant alterations in their concentration in serum.  Four occur with high frequency in some population groups and thus, can be considered as polymorphic.  TBG-Poly (Fig. 1), with no alterations of its physical or biological properties, has been detected in 16% and 20% of the French Canadian and Japanese populations, respectively (26, 57).  TBG-S exhibits a slower mobility on polyacrylamide gel electrophoresis and cathodal shift on IEF (54, 55), owing to the loss of a negative change due to the replacement of the normal Asp171 by Asn (56) (Figs. 1 and 4). It has an allele frequency of 5 to 16% in Black populations of African origin and 2 to 10% in Pacific Islanders.  The molecular structure of two other polymorphic TBG variants has not been identified.  TBG-F has an allele frequency of 3.2% in Eskimos residing on the Kodiac and St. Lawrence islands.  It has a slight anodal (fast) mobility on IEF (71).  TBG-C1 has been identified in subjects inhabiting two Mali village (72).  It has a small cathodal shift on IEF and an allele frequency of 5.1%.  TBG-Cgo, resistant to high temperatures (59), has normal affinity for T4 and T3.  All SERPINs except human TBG have a Phe at a position corresponding to Tyr309.  Structure modeling suggests that the replacement of the normal Tyr309 by Phe in TBG-Cgo, ties the internal α-helix hI1 to the molecule, thus stabilizing its tertiary structure (58).  Studies using recombinant TBG–Cgo showed that the molecule exists in loop expelled conformation.  However, when exposed at 37°C, the protein readily converts to a more stable loop inserted conformation explaining its subsequent  enhanced heat stability, as observed in vivo (73).

 

Figure 4. Microheterogeneity of TBG. Tracer amounts of l25I T4 were added to serum prior to submission to isoelectric focusing and radioautography. TBG C (common type) exhibits 6 bands spanning from pH 4.18 to 4.58. Three of the six are major and shown here between pH 4.35 and 4.50. TBG-Slow (TBG S) from a hemizygous male shows a cathodally shifted pattern. A mixed pattern occurs in heterozygous females expressing both TBG-C and TBG-S. [Reproduced from Waltz et al (56)].

 

Biological Consequences of Structural Changes Caused by Mutations in the TBG Gene

 

The mechanisms whereby structural abnormalities of the TBG molecule produce the variant phenotypes have been investigated by expression of some of these molecules in living cells.  Contrary to earlier speculation, increased extracellular degradation due to instability is a rare cause reduced concentration of the variant TBG in serum (38).  More commonly, intracellular retention and degradation of the defective TBG molecules is responsible for their presence in low concentrations in serum (45, 51, 60, 74).  Of note is the full intracellular retention of TBG-CD5 despite synthesis in normal quantities.  A single amino acid substitution in TBG-CD5 is sufficient to alter its tertiary structure and thus prevent export.  The same finding in the case of TBG-CDJ has been traced to its retention within the endoplasmic reticulum.  Furthermore, the increased amount of GRP78 mRNA in cells transfected with TBG-PDJ suggests that association of this TBG variant with the GRP78 molecular chaperon is responsible for its impaired secretion (51).  The variant TBG-AL is unique and important as it provides information about the function of the signal peptide. The resulting variable decrease in the serum TBG concentration associated with diminished in vitro secretion is compatible with impaired cotranslational processing (36). 

 

Several speculations regarding the properties of variant TBGs have been confirmed based on the elucidation of the TBG structure by X-ray crystallography (75). The reduced ligand-binding of TBG-SD (38, 39) can be explained by the direct proximity of the amino acid substitution to the binding pocket.  Indeed, the methyl group of the side chain of Thr23, replacing the normal Ser, will sterically hinder the binding of T4.  Similarly, in TBG-A, the replacement of Ala191 by Thr (47) perturbs the H-bounds that stabilizes the binding pocket, leading to the reduced T4 binding.  In contrast, the loss of His331 in TBG-Q (H331Y) (44, 49) allows unrestricted loop insertion in the upper half of the A-sheet, accounting for the increased in serum denatured (dn)TBG and reduced T4 binding.

 

TBG deficiency was found to coexist in the same family with resistance to thyroid hormone beta (RTHß) (76). Both TBG (P50fs51X) and THRB (P453A) gene mutations have been previously described in unrelated families (16, 77) but not in the same family. The mother harbored both gene mutations, whereas the proband and his sister had only the THRB gene mutation and a brother only the TBG gene mutation. This family illustrates the difficulty that might be encountered in the interpretation of thyroid function tests when different genetic defects, having opposite effect on thyroid function tests, coexist in the same family, and especially the same individual.

 

TRANSTHYRETIN (TTR) DEFECTS

 

Sequencing of the TTR gene, formerly known as thyroxine-binding prealbumin (TBPA) on chromosome 18 (18q11.2-q12.1), has uncovered mutations that produce variant TTR molecules with or without alterations in the binding affinity for iodothyronines (2, 78).  Only those known to affect iodothyronine binding are listed in table 2.  Some of the TTR variants are responsible for the dominantly inherited familial amyloidotic polyneuropathy (FAP), causing multiple organ failure and death in early adulthood.(78).  Because TTR has a relatively lower affinity for T4 (about 100-fold lesser than that of TBG), it plays a minor role in thyroid hormone transport in blood.  Accordingly, changes in the TTR concentration in serum and variant TTRs with reduced affinity for T4 have little effect on the concentration of serum T4 (79, 91).  Only variant TTRs with a substantially increased affinity for iodothyronines produce significant elevation in serum T4 and rT3 concentrations and account for 2% of subjects with euthyroid hyperthyroxinemia (90).

 

Table 2.  TTR Variants with Altered Affinity for T4 and Potentially an Effect on Tests of Thyroid Function in Serum

AFFINITY FOR T4

Mutant / Normal

TTR

CONCENTRATION

CODON

Number

AMINO ACID

(Normal - Variant)

REFERENCES

 

HOMO*

HETERO*

 

DECREASED

<0.1

0.17 - 0.41

N

30

Val - Met

(79, 80)

 

0.54

 

58

Leu - His

(80)

 

0.45

 

77

Ser - Tyr

(80)

 

0.19 – 0.46

N

84

Ile - Ser

(79, 80)

~1.0

0.44

 

 

Val - Ile

(80)

INCREASED

 

 

3.5†

N

6

Gly - Ser

(81-83)

8.3-9.8

3.2 - 4.1‡

N

109

Ala - Thr

(80, 84-86)

 

 

N

109

Ala - Val

(85)

 

 

Inc or N

119

Thr - Met

(87-90)

*  HOMO, homozygous; HETERO, heterozygous.

† Probably overestimated since the subjects harboring this TTR variant have normal serum TT4 concentrations.

‡  Affinity of recombinant TTR Thr109 is 9-fold that of the normal TTR (86).

Variant TTR tested and shown not to have altered affinity to T4 are: Ala60, (hetero) (79, 80).

N, normal; Inc, increased

Endonucleases useful in the identification of TTR variants: Msp I -ve for Ser6 in exon 2 associated PHA; BsoFI -ve and Fnu 4H +ve for Thr109; BsoFI -ve for Val109 and Nco I +ve for Met119, all in exon 4.

 

A family with elevated total T4 concentration which was predominantly bound to TTR was first described in 1982 by Moses et al (92).  The inheritance was autosomal dominant and affected members were clinically euthyroid with normal free T4 levels measured by equilibrium dialysis.  The variant TTR has a single nucleotide substitution replacing the normal Ala109 with a Thr which increases its affinity for T4, rT3 and tetraiodothyroacetic acid and to a lesser extend T3 and triiodothyroacetic acid (84, 86).  Crystallographic analysis of this variant TTR revealed an alteration in the size of the T4-binding pocket (93).  Another TTR gene mutation involving the same codon has been subsequently described (85).  This mutant TTR with Val109 has an increased affinity for T4 that is of similar magnitude as TTR Thr109, about 10-fold higher than that of wild-type TTR.

 

A more common defect found in subjects with prealbumin associated hyperthyroxinemia (PAH) is a point mutation in exon 4 of the TTR gene replacing the normal Thr119 with Met (90).  First described in a single individual with normal serum total and free T4 levels (89), the majority of subsequently identified heterozygous subjects harboring the TTR Met119 had an increase in the fraction of T4 and rT3 associated with TTR, but only few had serum T4 levels above the upper limit of normal.  Furthermore, their hyperthyroxinemia appears to be transient, usually in association with non-thyroidal illness  (90).  The variant TTRs associated with PAH are not amyloidogenic.

 

The unique occurrence in an Argentinian family of TTR A109T, known to have increased affinity for T4, in association with TGB-PD A64D mitigated the phenotype of the latter mutation (42)

 

Variant TTRs without Known Biological Effects

 

Several TTR variants have been found that do not alter the properties of the molecule, nor cause FAP, and are thus of no clinical significance.  Of interest is a TTR variant found in the rhesus monkey, Macaca mulatta, but not in man (94, 95)[.  This variant has a slower electrophoretic mobility resulting in three phenotypes which exhibit: (a) a single rapidly migrating band similar to that found in human and other primates (PAFF); (b) a single slowly migrating band cathodal to albumin (PASS); and (c) a five banded form corresponding to the various tetrameric recombinants present in the heterozygous state possessing the two different subunits (PAFS).  This finding was important because the variant rhesus PA-S could be hybridized in vitro with human TTR yielding a five-banded pattern hence, demonstrating that human TTR is also a tetramer.  All naturally occurring and hybrid polymorphic variants show no detectable alteration in the binding of either T4 or retinol binding protein  (96).

 

HUMAN SERUM ALBUMIN (HSA) DEFECTS

 

Another form of dominantly inherited euthyroid hyperthyroxinemia, later to be linked to the albumin gene on chromosome 4 (4q11-q13), was first described in 1979 (97, 98).  Known as familial dysalbuminemic hyperthyroxinemia (FDH) (99), it is the most common cause of inherited increase in total T4 in serum in the Caucasian population, producing on the average a 2-fold increase in the serum total T4 concentration.  In a study of 430 subjects suspected of having euthyroid hyperthyroxinemia 12% were proven to have FDH (90).  The prevalence varies from 0.01 to 1.8%, depending on the ethnic origin, with the highest prevalence in Hispanics (100-103).  This form of FDH has not been reported in subjects of African origin and the isolated occurrence in a Chinese (104) was possibly brought by Hispanic travelers (see below).  The euthyroid status of subjects with FDH has been confirmed by normal TSH response to TRH, normal free T4 concentration measured by equilibrium dialysis using appropriate buffer systems, normal T4 production rate and normal serum sex hormone-binding globulin concentration (97, 99, 105, 106).  Nevertheless, the falsely elevated free T4 values, when estimated by standard clinical laboratory techniques, has often resulted in inappropriate thyroid gland ablative or drug therapy (107-109).  A recent survey of commonly used commercial tests for measurement of free T4 indicates that equilibrium or symmetric dialysis are the only tests that will consistently provide accurate values in subjects with FDH (110), in particular one using dialysis in combination with tandem mass spectrometry (111).

 

FDH is suspected when serum total T4 concentration is increased without proportional elevation in total T3 level and non-suppressed serum TSH.  Half of affected subjects have also rT3 values above the normal range (112) (Table 3). Since the same combination of test results are found in subjects with the Thr109 TTR variant, the diagnosis of FDH should be confirmed by the demonstration that an increased proportion of the total serum T4 migrates with HSA on non-denaturing electrophoresis or precipitates with anti-HSA serum.

 

Table 3.  Albumin Variants with Increased Affinities for Iodothyronines, Their Effect on the Serum Concentrations of and Affinities to these Hormones

 

VARIANT

SERUM CONCENTRATION

 

 

BINDING AFFINITY (Ka)

of the variant albumins

Reference

T4

µg/dl

T3

ng/dl

rT3

ng/dl

N

T4

T3

 

 

(Fold the normal mean)

 

(Fold the normal mean)

WT

8.0 ± 0.2

125 ± 4

22.5 ± 0.9

83

1

1

A

R218H

16.0 ± 0.5

(2.0)

154 ± 3

(1.2)

33.1 ± 1.1

(1.5)

83

(10 – 15)

(4)

a,(113, 114)

R218P

135 ± 17

(16.8)

241 ± 25

(1.9)

136 ± 13

(6.1)

8

(11-13*)

(1.1*)

(9, 10)

R218S

70

(8.8)

159

(1.3)

55.7

(2.6)

1

NM

NM

(11)

R222I

21±1.4

(2.6)

135±18

(1.2)

1417±107

(86)

8

NM

NM

a, (115)

L66P

8.7

(1.1)

320

(3.3)

22.3

(1)

6

(1.5)

(40)

(3)

Values reported are means ± standard error, and the number of subjects per genotype are indicated under ‘‘N.’’

* Determined at saturation. Affinities are higher at the concentrations of T4 and T3 found in serum.

NM, not measured

a, Personal observation

All data were generated in the Chicago laboratory except for 4 of the 8 individuals with ALB R218P and hose with ALB R222I, provided by Nadia Schoenmakers, University of Cambridge, UK.

 

A tight linkage between FDH and the HSA gene (lod score 5.25) was found in a large Swiss-Amish family using two polymorphic markers (112).  This was followed by the identification of a missense mutation in codon 218 of the HSA gene replacing the normal arginine with a histidine (R218H) (113, 116).  Furthermore, the same mutation was present in all subjects with FDH from 11 unrelated families.  Its association with a Sac I+ polymorphism, suggest a founder effect and is compatible with ethnic predilection of FDH (113).  The coexistence of FDH and a TTR variant with increased affinity for T4 in the same individual (82, 83) and FDH with TBG-PD in another (117) have been reported.  In both instances these individuals were the product of parents each heterozygous for of one of the two defects.

 

Another mutation in codon 218, with increased affinity to iodothyronines, was first identified by Wada et al (9).  The mutation, a replacement of the normal Arg218 with a Pro (R218P), initially believed to be unique for Japanese was also identified in a Swiss family with no Asian ancestry (Fig. 5) (10).  In this form of FDH, serum total T4 levels are 14-20-fold the normal mean, a level confirmed by measurements in serum extracts by HPLC.  Total rT3 and T3 concentrations are 7- and 2-fold above the mean, respectively.  Thus, in order to maintain a normal free T4 level, the calculated affinity constant (Ka) of HSA R218P should be about 16-fold higher than that of HSA R218H.  Surprisingly, the Kas measured at saturation were similar, 5.4 x 106 M-1 and 6.4 x 106 M-1 for HSA R218H, respectively (10, 118, 119) (Table 3).  However, at T4 concentrations equivalent to those found in subjects with HSA R218P, the dialyzable FT4 concentration was 11-fold higher in serum of subjects with HSA R218H and 49-fold higher in serum with the common type HSA only (10).

 

Figure 5. A Swiss family with HSA R218P: genotyping, pedigree and thyroid function tests. A, Genotyping for the mutation HSA R218P. Results are aligned with each subject depicted on the pedigree in B. Amplification of a segment of the HSA gene containing the mutation with a mismatched oligonucleotide primer creates a new restriction site for Ava II only in the presence of the mutant nucleotide (CGC -> CCC). Affected subjects expressing proline 218 (CCC) show a 122 bp DNA fragment produced by enzymatic digestion of the mutant allele. Note that all affected subjects are heterozygous and that the 153 bp fragment amplified from DNA of the two normal subjects, expressing arginine 218 (CGC) only, resists enzymatic digestion. B, Pedigree of the family. Roman numerals indicate each generation and numbers below each symbol identify the subject. Individuals expressing the FDH phenotype are indicated by half-filled symbols. C, Thyroid function tests. Results are aligned with each symbol. Values outside the normal range are in bold numbers. Note the disproportionate increase in serum T4 concentration as compared to that of T3 in the affected individuals. Subject I-1, a year old man, had diabetes mellitus with multiple organ complications and mild subclinical hypothyroidism, explaining the relatively lower serum T4 and T3 but not rT3 levels. [Adapted from Pannain et al (10)].

 

More recently two additional HSA gene mutations have been identified.  One in the same codon resulting in a different amino acid substitution (R218S) (11) and another in a different amino acid (R222I) (115) in the proximity of the same iodothyronine-binding pocket (Fig. 6).  While both manifest increased affinity for T4 and rT3, it is considerably higher for T4 in the former and for rT3 in the latter (Table 3). It is of note that the two amino acids, 218 and 222, involved in the gain-of-function mutations are located in the main predominantly hydrophobic pocket where T4 is bound in a cisoid conformation (120).

 

Figure 6. The structures of HSA in the presence of T4 as modeled on the structures 1BM0, 1HK1, 1HK3 in the Protein Data Bank (http://www.rcsb.org/pdb/home/home.do). Top panel shows on the left the entire WT HSA molecule (in green) with its four T4 binding sites [T4(1) to T4(4)] according to Petitpas et al (120) and to the right a close up of the binding pocket, T4 (1) containing arginine’s 218 and 222 along with the T4 molecule (carbons are in white, nitrogen’s in blue, oxygens in red and iodine in magenta). In the bottom panel are represented the structures of the T4 (1) binding pockets of the four mutant HSA showing, a better accommodation of T4 than in the WT HSA and thus, resulting in enhanced binding (From Erik Schoenmakers, University of Cambridge, UK).

 

A fifth gain-of-function mutation, a replacement of the normal Leu66 with a Pro (L66P) has been identified in a single Thai family (3).  It produces a 40-fold increase in the affinity for T3 but only 1.5-fold increase in the affinity for T4 (Table 3).  As a consequence, patients have hypertriiodothyroninemia but not hyperthyroxinemia.  In this FDH-T3, serum T3 concentrations are falsely low, or even undetectable, when T3 is measured using an analog of T3 as a tracer rather than a radioisotope.  It has resulted in the inappropriate treatment with thyroid hormone (3).

 

Bisalbuminemia and Analbuminemia

 

Variant albumins, with altered electrophoretic mobility produce "bisalbuminemia" in the heterozygotes (121).  T4 binding has been studied in subjects from unrelated families with a slow HSA variant.  In two studies only the slow moving HSA bound T4 (122, 123) and in another, both (124).  The differential binding of T4 to one of the components of bisalbumin may be due to enhanced binding to the variant component with charged amino acid sequence.  Bisalbuminemia does not seem to be associated with gross alterations in thyroid hormone concentration in serum.

 

Analbuminemia is extremely rare, occurring in less than 1 in a million individuals (125).  The first case was reported in 1954 (126) but the HSA gene mutation was identified 56 years later (127).  The less than 50 cases so far reported have nonsense mutations causing premature termination of translation or splicing defects (128).  Despite the complete lack of such an important substance, symptoms are remarkably mild owing to the compensation by an increase in non-albumin serum proteins.  Studies with respect to T4-transport showed no clear effect or slight increase total serum iodothyronines, associated with increased levels of TBG and TTR. (128, 129).  The latter two normalized when serum HSA was restored to normal by multiple transfusions (129).

 

ACKNOWLEDGMENTS

 

Supported in part by grants DK-15070 from the National Institutes of Health (USA).

 

REFERENCES

 

  1. Beierwaltes WH, Robbins J 1959 Familial increase in the thyroxine-binding sites in serum alpha globulin. J Clin Invest 38:1683-1688.
  2. Pappa T, Ferrara AM, Refetoff S 2015 Inherited defects of thyroxine-binding proteins. Best Pract Res Clin Endocrinol Metab 29:735-747.
  3. Sunthornthepvarakul T, Likitmaskul S, Ngowngarmratana S, Angsusingha K, Sureerat K, Scherberg NH, Refetoff S 1998 Familial dysalbuminemic hypertriiodothyroninemia: a new dominantly inherited albumin defect. J Clin Endocrinol Metab 83:1448-1454.
  4. Refetoff S 1989 Inherited thyroxine-binding globulin (TBG) abnormalities in man. Endocr Rev 10:275-293.
  5. Refetoff S, Robin NI, Alper CA 1972 Study of four new kindreds with inherited thyroxine-binding globulin abnormalities: Possible mutations of a single gene locus. J Clin Invest 51:848-867.
  6. Burr WA, Ramsden DB, Hoffenberg R 1980 Hereditary abnormalities of thyroxine-binding globulin concentration. Quart J Med 49:295-313.
  7. Trent JM, Flink IL, Morkin E, Van TuinenP, Ledbetter  DH 1987 Localization of the human thyroxine-binding globulin gene to the long arm of the X chromosome (Xq21-22). Am J Hum Genet 41:428-435.
  8. Mori Y, Miura Y, Oiso Y, Seo H, Takazumi K 1995 Precise localization of the human thyroxine-binding globulin gene to chromosome Xq22.2 by fluorescence in situ hybridization. Hum Genet 96:481-482.
  9. Wada N, H. C, Shimizu C, Kijima H, Kubo M, Koike T 1997 A novel misssense mutation in codon 218 of the albumin gene in a distinct phenotype of familial dysalbuminemic hyperthyroxinemia in a Japanese kindred. J Clin Endocriol Metab 82:3246-3250.
  10. Pannain S, Feldman M, Eiholzer U, Weiss RE, Scherberg NH, Refetoff S 2000 Familial dysalbuminemic hyperthyroxinemia in a Swiss family caused by a mutant albumin (R218P) shows an apparent discrepancy between serum concentration and affinity for thyroxine. J Clin Endocrinol Metab 85:2786-2792.
  11. Greenberg SM, Ferrara AM, Nicholas ES, Dumitrescu AM, Cody V, Weiss RE, Refetoff S 2014 A Novel Mutation in the Albumin Gene (R218S) Causing Familial Dysalbuminemic Hyperthyroxinemia in a Family of Bangladeshi Extraction. Thyroid 24:945-950.
  12. Okamoto H, Mori Y, Tani Y, Nakagomi Y, Sano T, Ohyama K, Saito H, Oiso Y 1996 Molecular analysis of females manifesting thyroxine-binding globulin (TBG) deficiency: selective X-chromosome inactivation responsible for the difference between phenotype and genotype in TBG-deficient females. J Clin Endocrinol Metab 81:2204-2208.
  13. Gawandi S, Jothivel K, Kulkarni S 2022 Identification of a novel mutation in thyroxine-binding globulin (TBG) gene associated with TBG-deficiency and its effect on the thyroid function. J Endocrinol Invest 45:731-739.
  14. Refetoff S, Selenkow HA 1968 Familial thyroxine-binding globulin deficiency in a patient with Turner's syndrome (X0): Genetic study of a kindred. N Engl J Med 278:1081-1087.
  15. Reutrakul S, Janssen OE, Refetoff S 2001 Three novel mutations causing complete T4-binding globulin deficiency. J Clin Endocrinol Metab 86:5039-5044.
  16. Mannavola D, Vannucchi G, Fugazzola L, Cirello V, Campi I, Radetti G, Persani L, Refetoff S, Beck-Peccoz P 2006 TBG deficiency: description of two novel mutations associated with complete TBG deficiency and review of the literature. J Mol Med 84:864-871.
  17. Moeller LC, Appiagyei-Dankah Y, Kohler B, Biebermann H, Janssen OE, Fuhrer D 2015 Two Novel Mutations in the Serpina7 Gene Are Associated with Complete Deficiency of Thyroxine-Binding Globulin. Eur Thyroid J 4:108-112.
  18. Domingues R, Bugalho MJ, Garrão A, Boavida JM, Sobrinho L 2002 Two novel variants in thyroxine-binding globulin (TBG) gene behind the diagnosis of TBG deficiency. Eur J Endocrinol 146:485-490.
  19. Ueta Y, Mitani Y, Yoshida A, Taniguchi S, Mori A, Hattori K, Hisatome I, Manabe I, Takeda K, Sato R, Ahmmed GU, Tsuboi M, Ohtahara A, Hiroe K, Tanaka Y, Shigemasa C 1997 A novel mutation causing complete deficiency of thyroxine binding globulin. Clin Endocrinol (Oxf) 47:1-5.
  20. Hershkovitz E, Leiberman E, Refetoff S, Pilpell D, Phillip M 1995 High prevalence of thyroxine-binding globulin deficiency among Bedouin infants in southern Israel. Isr J Med Sci 31:500-502.
  21. Miura Y, Hershkovitz E, Inagaki A, Parvari R, Oiso Y, Phillip M 2000 A novel mutation causing complete thyroxine-binding globulin deficiency (TBG-CD-Negev) among the Bedouins in southern Israel. J Clin Endocrinol Metab 85:3687-3689.
  22. Su CC, Wu YC, Chiu CY, Won JG, Jap TS 2003 Two novel mutations in the gene encoding thyroxine-binding globulin (TBG) as a cause of complete TBG deficiency in Taiwan. Clin Endocrinol (Oxf) 58:409-414.
  23. Li P, Janssen OE, Takeda K, Bertenshaw RH, Refetoff S 1991 Complete thyroxine-binding globulin (TBG) deficiency caused by a single nucleotide deletion in the TBG gene. Metabolism 40:1231-1234.
  24. Carvalho GA, Weiss RE, Refetoff S 1998 Complete thyroxine-binding globulin (TBG) deficiency produced by a mutation in the acceptor splice site causing frameshift and early termination of translation (TBG-Kankakee). J Clin Endocrinol Metab 83:3604-3608.
  25. Lacka K, Nizankowska T, Ogrodowicz A, Lacki JK 2007 A Novel Mutation (del 1711 G) in the TBG Gene as a Cause of Complete TBG Deficiency. Thyroid 17:1143-1146.
  26. Mori Y, Takeda K, Charbonneau M, Refetoff S 1990 Replacement of Leu227 by Pro in thyroxine-binding globulin (TBG) is associated with complete TBG deficiency in three of eight families with this inherited defect. J Clin Endocrinol Metab 70:804-809.
  27. Domingues R, Font P, Sobrinho L, Bugalho MJ 2009 A novel variant in Serpina7 gene in a family with thyroxine-binding globulin deficiency. Endocrine 36:83-86.
  28. Personal-Observation.
  29. Carvalho GA, Weiss RE, Vladutiu AO, Refetoff S 1998 Complete deficiency of thyroxine-binding globulin (TBG-CD Buffalo) caused by a new nonsense mutation in the thyroxine-binding globulin gene. Thyroid 8:161-165.
  30. Reutrakul S, Dumitrescu A, Macchia P, Moll GWJ, Vierhapper H, Refetoff S 2002 Complete thyroxine-binding globulin (TBG) deficiency in two families without mutations in coding or promoter regions of the TBG gene: in vitro demonstration of exon skipping. J Clin Endocrinol Metab 87:1045-1051.
  31. Yamamori I, Mori Y, Seo H, Hirooka Y, Imamura S, Miura Y, Matsui N, Oiso Y 1991 Nucleotide deletion resulting in frameshift as a possible cause of complete thyroxine-binding globulin deficiency in six Japanese families. J Clin Endocrinol Metab 73:262-267.
  32. Yamamori I, Mori Y, Miura Y, Tani Y, Imamura S, Oiso Y, Seo H 1993 Gene screening of 23 Japanese families with complete thyroxine-binding globulin deficiency: Identification of a nucleotide deletion at codon 352 as a common cause. Endocr J 40:563-569.
  33. Yorifuji T, Muroi J, Uematsu A, Momoi T, Furusho K 1998 Identification of a novel variant of the thyroxine-binding globulin (TBG) in a Japanese patient with TBG deficiency (abstract). Horm Res 50 (suppl 3):68.
  34. Murata Y, Takamatsu J, Refetoff S 1986 Inherited abnormality of thyroxine-binding globulin with no demonstrable thyroxine-binding activity and high serum levels of denatured thyroxine-binding globulin. N Engl J Med 314:694-699.
  35. Moeller LC, Fingerhut A, Lahner H, Grasberger H, Weimer B, Happ J, Mann K, Janssen OE 2006 C-Terminal Amino Acid Alteration rather than Late Termination Causes Complete Deficiency of Thyroxine-Binding Globulin CD-NeuIsenburg. J Clin Endocrinol Metab 91:3215-3218.
  36. Fingerhut A, Reutrakul S, Knuedeler SD, Moeller LC, Greenlee C, Refetoff S, Janssen OE 2004 Partial deficiency of thyroxine-binding globulin-allentown is due to a mutation in the signal Peptide. J Clin Endocrinol Metab 89:2477-2483.
  37. Bertenshaw R, Sarne D, Tornari J, Weinberg M, Refetoff S 1992 Sequencing of the variant thyroxine-binding globulin (TBG)-San Diego reveals two nucleotide substitutions. Biochim Biophys Acta 1139:307-310.
  38. Janssen OE, Astner ST, Grasberger H, Gunn SK, Refetoff S 2000 Identification of thyroxine-binding globulin-San Diego in a family from Houston and its characterization by in vitro expression using Xenopus oocytes. J Clin Endocrinol Metab 85:368-372.
  39. Sarne DH, Refetoff S, Nelson JC, Dussault J 1989 A new inherited abnormality of thyroxine-binding globulin (TBG-San Diego) with decreased affinity for thyroxine and triiodothyronine. J Clin Endocrinol Metab 68:114-119.
  40. Gomes Lima CJ, de Olivera Andrada M, Santos daCunha V, Linhares Maciel AAF, Lofrano-Porto A 2015 A novel variant in the SERPINA7 gene is associated with TBG partial deficiency in a woman and her two male siblings ENDO 2015, San Diego CA, FRI-068.
  41. Moeller LC, Vinzelberg P, Jaeger A, Fingerhut A, Mann K, Janssen OE 2008 Two novel mutations leading to partial and complete thyroxine-binding globulin deficiency Symposium of the German Society for Endocrinology, Salzburg, Austria

.

  1. Sklate RT, Olcese MC, Maccallini GC, Sarmiento RG, Targovnik HM, Rivolta CM 2014 Novel mutation p.A64D in the Serpina7 gene as a cause of partial thyroxine-binding globulin deficiency associated with increases affinity in transthyretin by a known p.A109T mutation in the TTR gene. Horm Metab Res 46:100-108.
  2. Mori Y, Seino S, Takeda K, Flink IL, Murata Y, Bell GI, Refetoff S 1989 A mutation causing reduced biological activity and stability of thyroxine-binding globulin probably as a result of abnormal glycosylation of the molecule. Mol Endocrinol 3:575-579.
  3. Takamatsu J, Refetoff S, Charbonneau M, Dussault JH 1987 Two new inherited defects of the thyroxine-binding globulin (TBG) molecule presenting as partial TBG deficiency. J Clin Invest 79:833-840.
  4. Janssen OE, Refetoff S 1992 In-vitro expression of thyroxine-binding globulin (TBG) variants: Impaired secretion of TBGPRO-227 but not TBGPRO-113. J Biol Chem 267:13998-14004.
  5. Refetoff S, Murata Y 1985 X-chromosome-linked inheritance of the variant thyroxine-binding globulin in serum of Australian Aborigines: Its physical, chemical and biological properties. J Clin Endocrinol Metab 60:356-360.
  6. Takeda K, Mori Y, Sobieszczyk S, Seo H, Dick M, Watson F, Flink IL, Seino S, Bell GI, Refetoff S 1989 Sequence of the variant thyroxine-binding globulin of Australian Aborigines: Only one of two amino acid replacements is responsible for its altered properties. J Clin Invest 83:1344-1348.
  7. Moeller LC, Edidin DV, Jaeger A, Mann K, Refetoff S 2010 A novel mutation leading to familial partial thyroxine-binding globulin deficiency (TBG Glencoe) 14th International Thyroid Congress, Paris, France.
  8. Bertenshaw R, Takeda K, Refetoff S 1991 Sequencing of the variant thyroxine-binding globulin (TBG)-Quebec reveals two nucleotide substitutions. Am J Hum Genet 48:741-744.
  9. Miura Y, Mori Y, Yamamori I, Tani Y, Murata Y, Yoshimoto M, Kinoshita E, Matsumoto T, Oiso Y, Seo H 1993 Sequence of a variant thyroxine-binding globulin (TBG) in a family with partial TBG deficiency in Japanese (TBG-PDJ). Endocr J 40:127-132.
  10. Miura Y, Mori Y, Kambe F, Tani Y, Oiso Y, Seo H 1994 Impaired intracellular transport contributes to partial thyroxine-binding globulin (TBG) deficiency in a Japanese family. J Clin Endocrinol Metab 79:740-744.
  11. Not Published.
  12. Ferrara AM, Pappa T, Fu J, Brown CD, Peterson A, Moeller LC, Wyne K, White KP, Pluzhnikov A, Trubetskoy V, Nobrega M, Weiss RE, Dumitrescu AM, Refetoff S 2015 A novel mechanism of inherited TBG deficiency: mutation in a liver-specific enhancer. J Clin Endocrinol Metab 100:E173-181.
  13. Daiger SP, Rummel DP, Wang L, Cavalli-Sforza LL 1981 Detection of genetic variation with radioactive ligands.IV.  X-linked, polymorphic genetic variation of thyroxin-binding globulin (TBG). Am J Hum Genet 33:640-648.
  14. Takamatsu J, Ando M, Weinberg M, Refetoff S 1986 Isoelectric focusing variant thyroxine-binding globulin (TBG-S) in American Blacks: Increased heat lability and reduced concentration in serum. J Clin Endocrinol Metab 63:80-87.
  15. Waltz MR, Pullman TN, Takeda K, Sobieszczyk P, Refetoff S 1990 Molecular basis for the properties of the thyroxine-binding globulin-slow variant in American Blacks. J Endocrinol Invest 13:343-349.
  16. Takeda K, Iyota K, Mori Y, Tamura Y, Suehiro T, Kubo Y, Refetoff S, Hashimoto K 1994 Gene screening in Japanese families with complete deficiency of thyroxine-binding globulin demonstrates that a nucleotide deletion at codon 352 may be a race specific mutation. Clin Endocrinol (Oxf) 40:221-226.
  17. Janssen EO, Chen B, Büttner C, Refetoff S, Scriba PC 1995 Molecular and structural characterization of the heat-resistant thyroxine-binding globulin-Chicago. J Biol Chem 270:28234-28238.
  18. Takamatsu J, Refetoff S 1986 Inherited heat stable variant thyroxine-binding globulin (TBG-Chicago). J Clin Endocrinol Metab 63:1140-1144.
  19. Kambe F, Seo H, Mori Y, Murata Y, Janssen OE, Refetoff S, Matsui N 1992 An additional carbohydrate chain in the variant thyroxine-binding globulin-Gary (TBGAsn-96) impairs its secretion. Mol Endocrinol 6:443-449.
  20. Murata Y, Refetoff S, Sarne DH, Dick M, Watson F 1985 Variant thyroxine-binding globulin in serum of Australian Aborigines: Its physical, chemical and biological properties. J Endocrinol Invest 8:225-232.
  21. Refetoff S, Dumont JE, Vassart G 2000 Thyroid disorders. In: Scriver CR, Beaudet AL, Sly WS, Valle D, Childs B, Vogeldstein B, (eds) The Metabolic and Molecular Basis of Inherited Disease.The Metabolic and Molecular Basis of Inherited Disease. 8 ed. Vol 3. MacGraw-Hill, New York, 4029-4075.
  22. Janssen OE, Bertenshaw R, Takeda K, Weiss R, Refetoff S 1992 Molecular basis of inherited thyroxine-binding globulin defects. Trends Endocrinol Metab 3:49-53.
  23. Sarne DH, Refetoff S, Murata Y, Dick M, Watson F 1985 Variant thyroxine-binding globulin in serum of Australian Aborigines. A comparison with familial TBG deficiency in Caucasians and American Blacks. J Endocrinol Invest 8:217-224.
  24. Kobayashi H, Sakurai A, Katai M, Hashizume K 1999 Autosomally transmited low concentration of thyroxine-binding globuline. Thyroid 9:159-163.
  25. Griffiths KD, Virdi NK, Rayner PHW, Green A 1985 Neonatal screening for congenital hypothyroidism by measurement of plasma thyroxine and thyroid stimulating hormone concentrations. Brit Med J 291:117-120.
  26. Brown SK, Bellisario R 1986 Measurement of thyroxine-binding globulin (TBG) levels from dried blood spot specimens: Detection of TBG deficiency and TBG excess in infants. In: Carter TP, Wiley AH, (eds) Genetic Disease: Screening and Management. Vol. Alan R. Liss, Inc., 373-384.
  27. Refetoff S, Charbonneau M, Sarne DH, Takamatsu J, Dussault JH 1989 Resistance to thyroid hormones and screening for high thyroxine at birth. Research for Congenital Hypothyroidism. Vol. Plenum Publishing Corp., 165-172.
  28. Hayashi Y, Mori Y, Janssen OE, Sunthornthepvarakul T, Weiss RE, Takeda K, Weinberg M, Seo H, Bell GI, Refetoff S 1993 Human thyroxine-binding globulin gene: Complete sequence and transcriptional regulation. Mol Endocrinol 7:1049-1060.
  29. Mori Y, Miura Y, Takeuchi H, Igarashi Y, Sugiura J, Oiso Y 1995 Gene amplification as a cause for inherited thyroxine-binding globulin excess in two Japanese families. J Clin Endocrinol Metab 80:3758-3762.
  30. Kamboh MI, Ferrell RE 1986 A sensitive immunoblotting technique to identify thyroxine-binding globulin protein heterogeneity after isoelectric focusing. Biochem Genet 24:273-280.
  31. Constans J, Ribouchon MT, Gouaillard C, Chaventré A, Clayton J 1992 A new polymorphism of thyroxin-binding globulin in three African groups (Mali) with endemic nodular goitre. Hum Genet 89:199-203.
  32. Duhan U, Patston P 2010 Explanation for the high heat stability of thyroxine binding globulin-Chicago. Endocr Regul 44:43-47.
  33. Miura Y, Kambe F, Yamamori I, Mori Y, Tani Y, Murata Y, Oiso Y, Seo H 1994 A truncated thyroxine-binding globulin due to a frameshift mutation is retained within the rough endoplasmic reticulum: A possible mechanism of complete thyroxine-binding globulin deficiency in Japanese. J Clin Endocrinol Metab 78:283-287.
  34. Zhou A, Wei Z, Read RJ, Carrell RW 2006 Structural mechanism for the carriage and release of thyroxine in the blood. Proc Natl Acad Sci U S A 103:13321-13326.
  35. Ferrara AM, Cakir M, Henry PH, Refetoff S 2013 Coexistence of THRB and TBG Gene Mutations in a Turkish Family. J Clin Endocrinol Metab 98:E1148-1151.
  36. Adams M, Matthews C, Collingwood TN, Tone Y, Beck-Peccoz P, Chatterjee KK 1994 Genetic analysis of 29 kindreds with generalized and pituitary resistance to thyroid hormone. Identification of thirteen novel mutations in the thyroid hormone receptor beta gene. J Clin Invest 94:506-515.
  37. Benson MD 1995 Amyloidosis (Chapter131). In: Scriver CR, Beaudet AL, Sly WS, Valle D, (eds) The Metabolic and Molecular Basis of Inherited Disease. Vol. McGraw Hill Inc., New York.
  38. Refetoff S, Dwulet FE, Benson MD 1986 Reduced affinity for thyroxine in two of three structural thyroxine-binding prealbumin variants associated with familial amyloidotic polyneuropathy. J Clin Endocrinol Metab 63:1432-1437.
  39. Rosen HN, Moses AC, Murrell JR, Liepnieks JJ, Benson MD 1993 Thyroxine interactions with transthyretin: a comparison of 10 naturally occurring human transthyretin variants. J Clin Endocrinol Metab 77:370-374.
  40. Akbari MT, Fitch NJ, Farmer M, Docherty K, Sheppard MC, Ramsden DB 1990 Thyroxine-binding prealbumin gene: a population study. Clin Endocrinol (Oxf) 33:155-160.
  41. Fitch NJS, Akbary MT, Ramsden DB 1991 An inherited non-amyloidogenic transthyretin variant, [Ser6]-TTR, with increased thyroxine-binding affinity, characterized by DNA sequencing. J Endocrinol 129:309-313.
  42. Lalloz MR, Byfield PG, Goel KM, Loudon MM, Thomson JA, Himsworth RL 1987 Hyperthyroxinemia due to the coexistence of two raised affinity thyroxine-binding proteins (albumin and prealbumin) in one family. J Clin Endocrinol Metab 64:346-352.
  43. Moses C, Rosen HN, Moller DE, Tsuzaki S, Haddow JE, Lawlor J, Liepnieks JJ, Nichols WC, Benson MD 1990 A point mutation in transthyretin increases affinity for thyroxine and produces euthyroid hyperthyroxinemia. J Clin Invest 86:2025-2033.
  44. Refetoff S, Marinov VSZ, Tunca H, Byrne MM, Sunthornthepvarakul T, Weiss RE 1996 A new family with hyperthyroxinemia due to transthyretin Val109 misdiagnosed as thyrotoxicosis and resistance to thyroid hormone. J Clin Endocrinol Metab 81:3335-3340.
  45. Rosen HN, Murrell JR, Liepnieks JL, Benson MD, Cody V, Moses AC 1994 Threonine for alanine substitution at position 109 of transthyretin differentially alters human transthyretin's affinity for iodothyronines. J Clin Endocrinol Metab 134:27-34.
  46. Alves IL, Divino CM, Schussler GC, Altland K, Almeida MR, Palha JA, Coelho T, Costa PP, Saraiva MJM 1993 Thyroxine binding in a TTR met 119 kindred. J Clin Endocrinol Metab 76:484-488.
  47. Curtis AL, Scrimshaw BL, Topliss DJ, Stockigt JR, George PM, Barlow JW 1994 Thyroxine binding by human transthyretin variants: mutations at position 119, but not 54, increase thyroxine binding affinity. J Clin Endocrinol Metab 78:459-462.
  48. Harrison HH, Gordon ED, Nichols WC, Benson MD 1991 Biochemical and clinical characterization of prealbuminCHICAGO:An apparently benign variant of serum prealbumin (transthyretin) discovered with high-resolution two-dimensional electrophoresis. Am J Med Gen 39:442-452.
  49. Scrimshaw BJ, Fellowes AP, Palmer BN, Croxson MS, Stockigt JR, George PM 1992 A novel variant of transthyretin (prealbumin), Thr119 to Met, associated with increased thyroxine binding. Thyroid 2:21-26.
  50. Woeber KA, Ingbar SH 1968 The contribution of thyroxine-binding prealbumin to the binding of thyroxine in human serum, as assessed by immunoadsorption. J Clin Invest 47:1710-1721.
  51. Moses AC, Lawlor J, Haddow J, Jackson IMD 1982 Familial euthyroid hyperthyroxinemia resulting from increased thyroxine binding to thyroxine-binding prealbumin. N Engl J Med 306:966-969.
  52. Steinrauf LK, Hamilton JA, Braden BC, Murrell JR, Benson MD 1993 X-ray crystal structure of the Ala-109 -> Thr variant of human transthyretin which produces euthyroid hyperthyroxinemia. J Biol Chem 268:2425-2430.
  53. Alper CA, Robin NI, Refetoff S 1969 Genetic polymorphism of Rhesus thyroxine-binding prealbumin: Evidence for tetrameric structure in primates. Proc Natl Acad Sci U S A 63:775-781.
  54. Bernstein RS, Robbins J, Rall JE 1970 Polymorphism of monkey thyroxine-binding prealbumin (TBPA): Mode of inheritance and hybridization. Endocrinol 86:383-390.
  55. van Jaarsveld PP, Branch WT, Edelhoch H, Robbins J 1973 Polymorphism of Rhesus monkey serum prealbumin.Molecular properties and binding of thyroxine and retinol-binding protein. J Biol Chem 248:4706-44712.
  56. Henneman G, Krenning EP, Otten M, Docter R, Bos G, Visser TJ 1979 Raised total thyroxine and free thyroxine index but normal free thyroxine. A serum abnormality due to inherited increased affinity of iodothyronines for serum binding protein. Lancet 1:639-642.
  57. Lee WNP, Golden MP, Van Herle AJ, Lippe BM, Kaplan SA 1979 Inherited abnormal thyroid hormone-binding protein causing selective increase of total serum thyroxine. J Clin Endocrinol Metab 49:292-299.
  58. Ruiz M, Rajatanavin R, Young RA, Taylor C, Brown R, Braverman LE, Ingbar S 1982 Familial dysalbuminemic hyperthyroxinemia: A syndrome that can be confused with thyrotoxicosis. N Engl J Med 306:635-639.
  59. DeCosimo DR, Fang SL, Braverman LE 1987 Prevalence of familial dysalbuminemic hyperthyroxinemia in Hispanics. Ann Intern Med 107:780-781.
  60. Jensen IW, Faber J 1988 Familial dysalbuminaemic hyperthyroxinemia: a review. J Royal Soc Med 81:34-37.
  61. Sapin R, Gasser F, Chambron J 1989 Hyperthyroxinémie familiale avec dysalbuminémie:Dépistage sur 21 000 patients a l'occasion d'un bilan thyroïden. Pathol Biol (Paris) 37:785-789.
  62. Arevalo G 1991 Prevalence of familial dysalbuminemic hyperthyroxinemia in serum samples received for thyroid testing. Clin Chem 37:1430-1431.
  63. Tang KT, Yang HJ, Choo KB, Lin HD, Fang SL, Braverman LE 1999 A point mutation in the albumin gene in a Chinese patient with familial dysalbuminemic hyperthyroxinemia. Eur J Endocrinol 141:374-378.
  64. DeNayer P, Malvaux P 1982 Hyperthyroxinemia associated with high thyroxine binding to albumin in euthyroid subjects. J Endocrinol Invest 5:383-386.
  65. Mendel CM, Cavalieri RR 1984 Thyroxine distribution and metabolism in familial dysalbuminemic hyperthyroxinemia. J Clin Endocrinol Metab 59:499-504.
  66. Fleming SJ, Applegate GF, Beardwell CG 1987 Familial dysalbuminemic hyperthyroxinemia. Postgrad Med J 63:273-275.
  67. Wood DF, Zalin AM, Ratcliffe WA, Sheppard MC 1987 Elevation of free thyroxine measurement in patients with thyrotoxicosis. Quart J Med 65:863-870.
  68. Croxson MS, Palmer BN, Holdaway IM, Frengley PA, Evans MC 1985 Detection of familial dysalbuminaemic hyperthyroxinaemia. Br Med J 290:1099-1102.
  69. Cartwright D, O'Shea P, Rajanayagam O, Agostini M, Barker P, Moran C, Macchia E, Pinchera A, John R, Agha A, Ross HA, Chatterjee VK, Halsall DJ 2009 Familial dysalbuminemic hyperthyroxinemia: a persistent diagnostic challenge. Clin Chem 55:1044-1046.
  70. Refetoff S, Scherberg NH, Yuan C, Wu W, Wu Z, McPhaul MJ 2020 Free Thyroxine Concentrations in Sera of Individuals with Familial Dysalbuminemic Hyperthyroxinemia: A Comparison of Three Methods of Measurement. Thyroid 30:37-41.
  71. Weiss RE, Angkeow P, Sunthornthepvarakul T, Marcus-Bagley D, Cox N, Alper CP, Refetoff S 1995 Linkage of familial dysalbuminemic hyperthyroxinemia to the albumin gene in a large Amish family. J Clin Endocrinol Metab 80:116-121.
  72. Sunthornthepvarakul T, Angkeow P, Weiss RE, Hayashi Y, Refetoff S 1994 A identiucal missense mutation in the albumin gene produces familial disalbuminemic hyperthyroxinemia in 8 unrelated families. Biochem Biophys Res Commun 202:781-787.
  73. Petersen CE, Scottolini AG, Cody LR, Mandel M, Reimer N, Bhagavan NV 1994 A point mutation in the human serum albumin gene results in familial dysalbuminaemic hyperthyroxinaemia. J Med Genet 31:355-359.
  74. Schoenmakers N, Moran C, Campi I, Agostini M, Bacon O, Rajanayagam O, Schwabe J, Bradbury S, Barrett T, Geoghegan F, Druce M, Beck-Peccoz P, O'Toole A, Clark P, Bignell M, Lyons G, Halsall D, Gurnell M, Chatterjee K 2014 A novel albumin gene mutation (R222I) in familial dysalbuminemic hyperthyroxinemia. J Clin Endocrinol Metab 99:E1381-1386.
  75. Petersen CE, Ha C-E, Jameson DM, Bhagavan NV 1996 Mutations in a specific human serum albumin thyroxine binding site define the structural basis of familial dysalbuminemic hyperthyroxinemia. J Biol Chem 271:19110-19117.
  76. Langsteger W, Stockigt JR, Docter R, Költringer P, Lorenz O, Eber O 1994 Familial disalbuminaemic hyperthyroxinaemia and inherited partial TBG deficiency: fist report. Clin Endocrinol (Oxf) 40:751-758.
  77. Petersen CE, Ha CE, Harohalli K, Park DS, Feix JB, Isozaki O, Bhagavan NV 1999 Structural investigations of a new familial dysalbuminemic hyperthyroxinemia genotype. Clin Chem 45:1248-1254.
  78. Petersen CE, Ha CE, Harohalli K, Park DS, Bhagavan NV 2000 Familial dysalbuminemic byperthyroxinemia may result in altered warfarin pharmacokinetics. Chem Biol Interact 124:161-172.
  79. Petitpas I, Petersen CE, Ha CE, Bhattacharya AA, Zunszain PA, Ghuman J, Bhagavan NV, Curry S 2003 Structural basis of albumin-thyroxine interactions and familial dysalbuminemic hyperthyroxinemia. Proc Natl Acad Sci USA 100:6440-6445.
  80. Peters T, Jr. 1985 Serum albumin. Adv Prot Chem 37:161-245.
  81. Sarcione EJ, Aungst CW 1962 Bisalbuminemia associated with albumin thyroxine-binding defect. Clin Chim Acta 7:297-298.
  82. Tarnoky AL, Lestas AN 1964 A new type of bisalbuminemia. Clin Chim Acta 9:551-558.
  83. Andreoli M, Robbins J 1962 Serum proteins and thyroxine-protein interaction in early human fetuses. J Clin Invest 41:1070-1077.
  84. Watkins S, Madison J, Galliano M, Minchiotti L, Putnam FW 1994 A nucleotide insertion and frameshift cause analbuminemia in an Italian family. Proc Natl Acad Sci U S A 91:2275-2279.
  85. Bennhold H, Peters H, Roth E 1954 Uber einen Fall von kompletter Analbuminaemie ohne wesentliche klinische Krankheitszichen. Verh Dtsch Gesellsch Inn Med 60:630-634.
  86. Ruhoff MS, Greene MW, Peters T 2010 Location of the mutation site in the first two reported cases of analbuminemia. Clin Biochem 43:525-527.
  87. Koot BG, Houwen R, Pot DJ, Nauta J 2004 Congenital analbuminaemia: biochemical and clinical implications. A case report and literature review. Eur J Pediatr 163:664-670.
  88. Hollander CS, Bernstein G, Oppenheimer JH 1968 Abnormalities of thyroxine binding in analbuminemia. J Clin Endocrinol Metab 28:1064-1066.