Archives

The Effect of Endocrine Disorders on Lipids and Lipoproteins

ABSTRACT

 

Endocrine disorders and the administration of various hormones can alter lipid metabolism and plasma lipid levels, which may increase or decrease the risk of atherosclerotic cardiovascular disease. In many instances the literature is not consistent with various studies reporting different results. These differences may be due to a variety of factors such as the differences in the severity of the disease state, differences in the duration of the disease, underlying genetic factors that differ between individuals and populations, differences in environmental factors such as diet, the presence of other abnormalities that can alter lipid metabolism such as obesity or diabetes, and other unrecognized factors that could influence the expression and manifestation of various endocrine disorders on lipid parameters. Prolactinomas are associated with an increase in total and LDL-C levels. GH deficient patients often have an increase in total cholesterol, LDL-C, and triglyceride levels and a decrease in HDL-C levels, whereas GH therapy decreases total cholesterol and LDL-C but increases Lp(a) levels. Acromegaly is associated with an increase in Lp(a) levels as seen in GH therapy, but paradoxically similar to GH deficiency, acromegaly is accompanied by an increase in plasma triglycerides and a decrease in HDL-C levels. Hypothyroidism leads to an increase in total cholesterol, LDL-C, and Lp(a) levels and normal or increased triglycerides and HDL-C. In contrast, hyperthyroidism is characterized by decreases in total cholesterol, LDL-C, and Lp(a) levels, as well as HDL-C levels. Patients with endogenous Cushing’s syndrome typically display an increase in total cholesterol and LDL-C, and triglycerides, while the administration of glucocorticoids frequently also increases HDL-C levels. Men with low testosterone levels may have high LDL-C and triglyceride levels and decreased HDL-C levels, although this relationship is confounded by obesity and the metabolic syndrome, a common cause of male hypogonadism. Androgen deprivation therapy results in an increase in LDL-C, triglycerides, and Lp(a) and a decrease in HDL-C. The effect of testosterone replacement therapy on plasma lipids and lipoproteins is modest and variable but high dose androgen therapy used by athletes can markedly decrease HDL-C and also reduce Lp(a) levels. The loss of estrogens (postmenopausal females) is associated with a modest increase in LDL-C with either no change or a small decrease in HDL-C. Estrogen administration decreases LDL-C and Lp(a) levels while increasing triglycerides and HDL-C levels but these effects are dependent on the dose and route of administration (transdermal has smaller effects than oral). Concurrent progesterone treatment has little or no effect on the decrease in LDL-C induced by estrogen administration but may blunt the estrogen effect on HDL-C and triglyceride levels depending on the androgenicity of the progesterone. The polycystic ovarian syndrome is associated with increases in LDL-C, triglycerides, and Lp(a) and decreases in HDL-C. The dyslipidemia that occurs with prolactinomas, GH deficiency, hypothyroidism, Cushing’s syndrome, male hypogonadism, androgen deprivation therapy, polycystic ovarian syndrome, and the loss of estrogens may contribute to an increased risk of atherosclerotic cardiovascular disease.

 

INTRODUCTION

 

Endocrine disorders and the administration of various hormones can alter lipid metabolism and plasma lipid levels, which may increase or decrease the risk of atherosclerotic cardiovascular disease (ASCVD). In this chapter we will discuss the effects of a number of endocrine disorders on lipid metabolism and plasma lipid and lipoprotein levels. It is worth noting that in many instances the literature is not consistent with various studies reporting different results. These differences may be due to a variety of factors such as the differences in the severity of the disease state, differences in the duration of the disease, underlying genetic factors that differ between individuals and populations, differences in environmental factors such as diet, the presence of other abnormalities that can alter lipid metabolism such as obesity or diabetes, and other unrecognized factors that could influence the expression and manifestation of various endocrine disorders on lipid parameters. In describing the alterations in lipid metabolism and plasma lipid and lipoprotein levels induced by various endocrine disorders we have tried to describe the typical alterations that have been most consistently observed, recognizing that these changes have not been observed in certain published reports and cannot be extrapolated to individual patients.

 

PROLACTINOMA

 

Effect of Prolactinomas on Lipid and Lipoprotein Levels

 

Most studies have shown that patients with a prolactinoma have modestly elevated plasma total cholesterol and LDL-C levels (1-8). In some studies plasma triglyceride levels are also elevated (1,2,4,8-10).  HDL-C levels have been reported to be decreased in some studies (7,8,10,11). Most studies have primarily included female patients with prolactinomas but dyslipidemia is also observed in men with hyperprolactinemia (4).  

 

Table 1. Effect of Hyperprolactinemia on Lipid and Lipoprotein Levels

Total Cholesterol

Increase

LDL-C

Increase

HDL-C

 No Change or Decrease

Triglycerides

No Change or Increase

 

The mechanisms accounting for the alterations in plasma lipid levels are not clear but could be related to a number of factors. First, prolactin may have direct effects on lipid metabolism. For example, prolactin decreases lipoprotein lipase activity in human adipose tissue and plasma lipoprotein lipase activity is decreased in patients with prolactinomas, which could increase triglyceride levels (2,12). Second, elevated prolactin levels are associated with decreased estrogen levels in women, a change that is associated with elevated LDL-C and decreased HDL-C levels. Third, elevated prolactin levels are associated with obesity, which could adversely affect plasma lipid levels (1). Finally, with large prolactinomas the secretion of growth hormone (GH) may be impaired, which can result in abnormal plasma lipid levels (2).

 

Lowering prolactin levels with dopamine agonists, such as bromocriptine or cabergoline, has been shown to decrease plasma total and LDL-C levels and in some instances triglycerides (1,6-8,13-19). However, it is unclear if this effect is solely due to lowering prolactin levels or to other effects of dopamine agonists. The administration of dopamine agonists to patients without prolactinomas has also been shown to induce changes in plasma lipid levels (20). It should be noted that in patients with very high prolactin levels (1355ug/L) pituitary surgery rapidly lowered prolactin levels (77ug/L) and a statistically significant decrease in total cholesterol and triglyceride levels was seen (21). LDL-C levels were also decreased 8.8% but didn’t achieve statistical significance perhaps due to the small number of patients studied (n=17). This observation suggests that lowering prolactin has beneficial effects on the lipid profile.  

 

Risk of Atherosclerotic Cardiovascular Disease (ASCVD)

 

In patients with prolactinomas, carotid-intima media thickness has been shown to be increased (9,10,22). Moreover, a positive association of serum prolactin concentrations with all-cause and cardiovascular mortality and events has been reported (23,24). This increase in cardiovascular mortality has been particularly noted in males with elevated prolactin levels (25,26). These results suggest that hyperprolactinemia might increase the risk of ASCVD. While prolactin induced abnormalities in lipids could contribute to this increased risk, it should be recognized that elevated prolactin levels also induce other metabolic abnormalities such as obesity, pro-inflammatory state, insulin resistance, and alterations in glucose metabolism that could accelerate atherosclerosis (8). 

 

GROWTH HORMONE DEFICIENCY

 

Effect of Growth Hormone Deficiency on Lipid and Lipoprotein Levels

 

Dyslipidemia is commonly observed in adults with growth hormone (GH) deficiency (27-34). Plasma total cholesterol, LDL-C, and triglyceride levels are elevated while HDL-C levels are decreased. Some studies reporting no difference in LDL size and others an increase in small dense LDL while Lp(a) levels in controls and in GH deficient patients are similar (30,31,35,36).  It should be recognized that GH deficiency leads to increased adiposity, which may be an important contributor to dyslipidemia (37). However, even when controlling for BMI, dyslipidemia is still present in GH deficient patients (27).

Table 2. Effect of Growth Hormone Deficiency on Lipid and Lipoprotein Levels

 

Total Cholesterol

Increase

LDL-C

Increase

HDL-C

Decrease

Triglycerides

Increase

Lp (a)

No change

 

 

Effect of Growth Hormone Therapy on Lipid and Lipoprotein Levels

 

Numerous studies have examined the effect of GH replacement therapy on serum lipid levels. A meta-analysis by Newman and colleagues reported on the effect of low dose GH replacement (<0.7mg/day; seven studies) and high dose GH replacement (>0.7mg/day; sixteen studies) involving over 1000 subjects (38). In both the low dose and high dose groups, GH replacement therapy decreased total and LDL-C levels but did not significantly affect either HDL-C or triglyceride levels. LDL-C levels were decreased by 11.3%. A meta-analysis of 37 trials by Maison et al also found that total and LDL-C levels were decreased with no significant changes in triglycerides or HDL-C by GH treatment (39). In a few studies, HDL-C levels have been observed to increase with GH therapy (32,40,41). For example, in a 15 year long term perspective study GH therapy reduced LDL-C and increased HDL-C levels, while having no significant effect on triglyceride levels (42). The ability of GH therapy to decrease LDL-C levels occurs even when patients are on statin therapy (43). Moreover, the decrease in LDL-C levels with GH treatment correlates with baseline LDL-C levels (i.e. the higher the LDL-C the greater the decrease with GH treatment) (44). Interestingly GH treatment increases Lp(a) levels (41,45-52). Of note, studies have shown that treatment with GH increases Lp(a) levels while treatment with IGF-1 decreases Lp(a) levels (53). Whether this increase in Lp(a) levels will enhance the risk of cardiovascular disease is unknown.

Table 3. Effect of Growth Hormone Therapy on Lipid and Lipoprotein Levels

Total Cholesterol

Decrease

LDL-C

Decrease

HDL-C

No Change or Increase

Triglycerides

No Change

Lp (a)

Increase

  

Mechanism for the Changes in Lipids and Lipoproteins in GH Deficiency

 

LDL-C

 

Studies have shown that GH increases the expression of hepatic LDL receptors (54,55). Additionally, GH decreases circulating PCSK9 levels, which would also increase hepatic LDL receptors (56). As a consequence, the clearance of LDL-C is accelerated by GH treatment (57,58). Thus, the increase in total cholesterol and LDL-C levels in GH deficient patients is likely due to a decrease in hepatic LDL receptors and therefore with GH administration the number of LDL receptors increases leading to a decrease in plasma LDL-C levels. Notably, in a patient with homozygous familial hypercholesterolemia, devoid of functional LDL receptors, GH treatment did not result in a decrease in LDL-C levels, whereas in GH deficient patients, normal subjects, and patients with heterozygous familial hypercholesterolemia treatment with GH resulted in a decrease in LDL-C levels (57). This observation further demonstrates the importance of LDL receptors in mediating the decrease in LDL-C levels in response to GH administration.

 

TRIGLYCERIDES

 

In GH deficient patients there is an increase in hepatic VLDL production and a reduction in VLDL clearance, which together could lead to an increase in plasma triglyceride levels (59). GH therapy stimulates VLDL secretion and increases VLDL clearance, which is likely due to its effects in up-regulating low density lipoprotein receptors, leading to a neutral effect on plasma triglyceride levels (60). The enhancement in VLDL secretion by GH treatment is likely facilitated by the well-recognized ability of GH to stimulate lipolysis in adipose tissue, which will provide fatty acids for the synthesis of triglycerides in the liver and enhance VLDL production (61).  GH increases fatty acid oxidation but this may not be able to offset the increased lipolysis and VLDL production (62). 

 

LIPOPROTEIN (a)

 

In transgenic mice expressing the human Apo (a) gene, GH administration increases the mRNA levels of Apo (a) and plasma levels of Apo (a) (63). The increased production of Apo (a) induced by GH could account for the increase in Lp(a) levels induced by GH treatment.

 

Risk of Cardiovascular Disease

 

Several observational studies have found that patients with hypopituitarism on conventional replacement therapy have an increased mortality that is primarily due to cardiovascular and cerebrovascular disease (64-68). Additionally, the risk of myocardial infarctions is increased in hypopituitarism (64,69). Moreover, increased coronary artery calcifications and carotid intima-media thickness have been observed in patients with GH deficiency (33,70-77). It is likely that the dyslipidemia that commonly occurs in GH deficient patients contributes to this increased risk of cardiovascular disease. However, GH deficient patients also display an increase in visceral adiposity, insulin resistance, impaired glucose metabolism, an increased prevalence of the metabolic syndrome, and an increased pro-inflammatory state with elevations in C-reactive protein and inflammatory cytokines, which could also contribute to an increased risk of cardiovascular disease (40). Since GH deficient patients have an increased risk of ASCVD one could consider GH deficiency as a risk enhancer when evaluating patients for lipid lowering therapy.

 

Whether treating GH deficient patients with GH replacement therapy reduces the risk of cardiovascular disease is uncertain, as there are no long-term randomized outcome studies. There are however a number of observational studies. Svensson and colleagues reported that in patients with hypopituitarism on GH replacement therapy the risk of myocardial infarctions was decreased but the occurrence of cerebrovascular events appeared to be increased compared to untreated patients (64). Bengtsson and colleagues reported that morbidity was not increased in patients with GH deficiency who were treated with GH compared to the general population and was even reduced compared to untreated patients (78). Holmer et al reported that in GH deficient patients, the risk of nonfatal stroke declined in males and females and nonfatal cardiac events decreased in males treated with GH replacement therapy (79). Finally, van Bunderen et al reported that GH deficient men receiving GH treatment had a mortality rate similar to the background population but women had an increase in cardiovascular mortality (80). Together these results suggest that GH therapy may reduce the risk of cardiovascular disease.

 

In non-randomized trials a decrease in carotid intima-media thickness was observed in GH deficient patients treated with GH (71,74,75,77,81-83). Other similar studies have not shown a decrease in carotid intima-media thickness with GH treatment [61]. Furthermore, in Brazilian patients with lifelong isolated GH deficiency, treatment with GH increased carotid intima-media thickness (84).

 

Thus, at this time it is uncertain whether GH replacement therapy will have beneficial effects on long term ASCVD outcomes. Randomized outcome trials will be required to definitively answer this question.

 

ACROMEGALY

 

Effect of Acromegaly on Lipid and Lipoprotein Levels

 

In patients with acromegaly an increase in plasma triglyceride levels and a decrease in HDL-C levels have been frequently observed (85-95). In one large retrospective study of 307 newly diagnosed patients with acromegaly, 33% of patients were noted to have elevated triglyceride levels (>150mg/dl) while 17% of men and 62% of women had low HDL-C levels defined by metabolic syndrome criteria (96). The effect of acromegaly on total cholesterol and LDL-C levels has been variable (85,86,88-91,93-95,97-100). However, an increase in small dense LDL levels and Apo B levels may be seen (92-94,101). Additionally, an increase in Lp(a) levels has been reported in several studies (94,99,102-104).

 

Table 4. Effect of Acromegaly on Lipid and Lipoprotein Levels

 

Total Cholesterol

Variable

LDL-C

Variable

HDL-C

Decrease

Triglycerides

Increase

Lp (a)

Increase

 

Treatment of acromegaly that normalizes GH and IGF-1 levels typically results in a decrease in plasma triglyceride levels and an increase in HDL-C levels (89,90,94,104-111). Additionally, small dense LDL and Lp(a) levels may also decrease (94,97,102-104,106-108). Interestingly, the GH-receptor antagonist, pegvisomant, increased TG levels in healthy men (112) and increased total and LDL-C levels and decreased Lp(a) levels in patients with acromegaly (97,98).

 

Mechanism for the Changes in Lipids and Lipoproteins in Acromegaly

 

TRIGLYCERIDES

 

The increase in plasma triglycerides has been shown to be associated with an increased triglyceride production rate (87). Treatment with GH stimulates VLDL secretion, which is likely facilitated by the ability of GH to enhance lipolysis that will provide fatty acids for the synthesis of triglycerides in the liver, thereby enhancing VLDL production (60,61). In addition, several studies have shown that lipoprotein lipase activity is decreased in patients with acromegaly, which could decrease the clearance of triglyceride rich lipoproteins (86,113,114). It is likely that the insulin resistance and abnormal glucose metabolism that frequently occurs in patients with acromegaly also contributes to the abnormalities in triglyceride metabolism.

 

HDL-C

 

LCAT, hepatic lipase, and phospholipid transfer protein have all been reported to be decreased in patients with acromegaly while some studies have shown a decrease in CETP and others an increase (93,95,101). Whether these changes account for the decrease in HDL-C levels is uncertain. A decrease in LCAT, CETP, and hepatic lipase could result in a decrease in reverse cholesterol transport (115).

 

LIPOPROTEIN (a)

 

In transgenic mice expressing the human Apo (a) gene, GH administration increases the mRNA levels of Apo (a) and plasma levels of Apo (a) (63). The increased production of Apo (a) induced by GH could account for the increase in Lp(a) levels in patients with acromegaly. Of note studies have shown that treatment with GH increases Lp(a) levels however treatment with IGF-1 decreases Lp(a) levels (53).

 

Risk of Cardiovascular Disease

 

Cardiovascular disease is increased in patients with acromegaly but much of this is related to acromegalic cardiomyopathy, valvular heart disease, and arrhythmias (116,117). It remains uncertain whether atherosclerotic cardiovascular disease is increased (116,117). A study using the German Acromegaly Registry did not observe an increase in myocardial infarctions or strokes in 479 patients with acromegaly compared to the general population (118). Similarly, a large cohort study from Korea with over 1800 patients with acromegaly also did not observe an increase in atherosclerotic cardiovascular disease events (119). Several studies have shown an increase in carotid intima-media thickness in patients with acromegaly (89,90,120-124). However, a study by Otsuki and colleagues showed that if one controls for risk factors carotid intima-media thickness in patients with acromegaly was similar to matched controls (125). In contrast, Ozkan and colleagues found that carotid intima-media thickness in patients with acromegaly was still increased even in matched controls (124). Several studies have shown that the treatment of acromegaly results in a decrease in carotid intima-media thickness (89,90,122,126). In contrast to the results seen in studies of carotid intima-media thickness, studies of coronary artery calcium score in patients with acromegaly have not consistently shown an increase in atherosclerosis. While Cannavo et al have shown an increase in coronary artery calcium, other studies have not shown an increase (127-130). In the study of Herrmann et al the coronary artery calcium score directly correlated with disease duration suggesting that patients with long standing acromegaly are more likely to develop atherosclerosis (131). Thus, whether acromegaly increases atherosclerosis and atherosclerotic cardiovascular disease events requires further investigation.

 

HYPOTHYROIDISM

 

Effect of Hypothyroidism on Lipid and Lipoprotein Levels

 

It has been recognized since the 1930s that hypothyroidism results in an increase in plasma cholesterol levels (132). Indeed, along with protein bound iodine, cholesterol levels were followed as a marker for treatment before immunoassays were developed for TSH and FT4.  The lipid profile of hypothyroid patients is characterized by an increase in total and LDL-C levels (132). LDL-C levels can be strikingly elevated, sometimes raising the suspicion of familial hypercholesterolemia. Hypothyroidism can also unmask familial dysbetalipoproteinemia (Type III hyperlipidemia) (133-135). In most studies there is not an increase in small dense LDL (132). It should be routine clinical practice to determine thyroid function in patients with significant elevations in LDL-C to rule out hypothyroidism as the cause of the hypercholesterolemia. The effect of hypothyroidism on HDL-C levels is variable with either no change or a modest increase in HDL-C levels but there is a more consistent increase in the concentration of HDL 2 particles (132,136). Similarly, hypothyroidism has either no effect or modestly increases plasma triglyceride levels (132). Of note, Lp(a) levels are also increased in hypothyroid patients (132,137-141).  In a study of 295 patients with overt hypothyroidism 56% had elevations in LDL-C, 34% had elevated LDL-C and elevated triglyceride levels, 1.5% had elevations only in triglycerides, and 8.5% had no lipid abnormalities (142). Patients with secondary hypothyroidism were more likely to have elevations in both LDL-C and triglyceride levels in this study (142). However, other studies have not observed a difference in the dyslipidemia in patients with primary or secondary hypothyroidism (143). In general, the changes in lipids and lipoprotein induced by hypothyroidism are pro-atherogenic and are more severe with severe hypothyroidism. Restoration of thyroid function improves the lipid abnormalities towards normal (132,142,144,145). A meta-analysis by Kotwal et al demonstrated that the treatment of hypothyroidism with levothyroxine resulted in a decrease in total cholesterol by -58 mg/dL (95% CI: -64.7, -52.1), LDL-C by -41 mg/dL (95% CI: -46.5, -35.7), HDL-C by -4.1 mg/dL (95% CI: -5.67, -2.61), triglycerides by -7.3 mg/dL (95% CI: -36.63, 17.87), apo A by -12.6 mg/dL (95% CI: -17.98, -7.19), apo B by -34.0 mg/dL (95% CI: 41.14, -26.77), and Lp(a) by -5.6 mg/dL (95% CI: -9.06, -2.14) (146).

 

Table 5. Effect of Hypothyroidism on Lipid and Lipoprotein Levels

 

 

Overt Hypothyroidism

Subclinical Hypothyroidism

Total Cholesterol

Increase

Normal to increased

LDL-C

Increase

Normal to increased

HDL-C

Normal to slightly increased

No change

Triglycerides

Normal to increase

Normal to increased

Lp(a)

Increase

No change

Apo B

Increase

Increase

Apo A-I

Increase

No change

 

Subclinical Hypothyroidism

 

The effects of subclinical hypothyroidism on lipid and lipoprotein levels have been highly variable with some studies showing changes similar to what is observed in patients with overt hypothyroidism and other studies showing no differences in patients with subclinical hypothyroidism compared to controls (147,148). These differences are likely related the types of patients included in the studies with variables such as age, ethnicity, duration of hypothyroid dysfunction, and the presence of other metabolic abnormalities such as insulin resistance (149). One key variable is the degree of thyroid dysfunction with studies that included patients with higher TSH levels (>10mIU/L) more likely to show that subclinical hypothyroidism is associated with abnormalities in lipid and lipoprotein levels (148).

 

An important issue in patients with subclinical hypothyroidism is whether one should treat with thyroid hormone replacement or just observe. Because of this uncertainty it has been of great interest to determine if the lipid profile in patients with subclinical hypothyroidism improves with thyroid hormone treatment. A large number of studies have explored this issue but the results have likewise been inconsistent with some studies showing potentially beneficial changes in the lipid profile and other studies showing no changes with treatment of subclinical hypothyroidism (147,148). A recent review also did not find firm evidence of a beneficial effect on the lipid profile with thyroid hormone treatment in patients with subclinical hypothyroidism (150). A meta-analysis by Kotwal et al demonstrated that the treatment of subclinical hypothyroidism with levothyroxine resulted in a decrease in total cholesterol by -12 mg/dL, LDL-C by -11 mg/dL, triglycerides by -4.5 mg/dL, apo B by -6.6 mg/dL, and Lp(a) by -1.99 mg/dL with no significant changes in HDL-C or apo AI (146). However, when this meta-analysis only included studies with either a placebo or observational control group they did not demonstrate any significant changes in lipids with levothyroxine therapy (146). It is likely that the patients with higher TSH levels and higher LDL-C levels will benefit from treatment with L-thyroxine (151).

 

Risk of Cardiovascular Disease in Subclinical Hypothyroidism

 

A major issue in patients with subclinical hypothyroidism is whether they are at increased risk of developing cardiovascular disease. Some but not all meta-analyses have suggested that subclinical hypothyroidism is associated with a small increase in cardiovascular risk particularly in young patients and patients whose TSH is greater than 10mIU/L (147,152-156). The length of time that a patient is hypothyroid and the degree of elevation of cholesterol may be important factors. Whether thyroid treatment lowers this risk is uncertain with some observational studies reporting a benefit and others reporting no benefit (147,148,157).  No randomized outcome studies have addressed whether treatment with thyroid hormone will reduce cardiovascular events in patients with subclinical hypothyroidism and without such studies it is difficult to be certain whether thyroid hormone replacement is indicated.

 

In patients with subclinical hypothyroidism carotid intima-media thickness (cIMT) is increased and two meta-analyses found that thyroid hormone treatment reduced cIMT suggesting a possible beneficial effect on atherosclerosis (158-160). This decrease in cIMT was associated with a reduction in plasma lipid levels. However, it should be noted that a recent randomized study of 185 subjects with subclinical hypothyroidism (TSH 6.35mIU/L) did not find any difference in cIMT after 18 months in the thyroid hormone  treated group compared to the placebo group (161). Only a small number of studies have examined coronary calcium scores but the limited data suggest an increase in coronary calcium in individuals with subclinical hypothyroidism (162-165).

 

It is recommended by the American Thyroid Association, and the American Association of Clinical Endocrinologists that subclinical hypothyroidism should be treated when the TSH level is >10 mIU/L (157).  Routine treatment for patients with TSH levels between 4.5 and 10mIU/L is not recommended but one can decide to initiate therapy based on individual factors, such as antibodies and symptoms (157). There are no recommendations by these societies to treat with thyroid hormone replacement for the purpose of correcting abnormal lipid and lipoprotein levels or reducing cardiovascular risk.  Since randomized clinical trials have not consistently shown a lipid-lowering benefit with thyroid hormone therapy in patients with subclinical hypothyroidism (TSH < 10mIU/L), patients with significant hyperlipidemia, should be treated with lifestyle changes and lipid-lowering medications.

 

Mechanism for the Changes in Lipids and Lipoproteins in Hypothyroidism

 

Thyroid hormone regulates the expression and activity of a number of key enzymes and receptors that regulate lipid and lipoprotein levels.

 

LDL-C

 

The primary mechanism by which hypothyroidism results in elevated total cholesterol and LDL-C levels is via a decrease in LDL receptor levels in the liver. Thyroid hormone stimulates the expression of LDL receptors and in hypothyroidism the number of hepatic LDL receptors is reduced leading to the decreased clearance of circulating LDL (132,166-171). This decreased clearance of LDL accounts for the increase in plasma LDL levels. Thyroid hormone stimulates LDL receptor expression by increasing SREBP-2 and/or by direct effects on the LDL receptor promoter (172,173). Finally, PCSK9 levels are increased with hypothyroidism, which could further contribute to a decrease in hepatic LDL receptor levels by accelerating the catabolism of LDL receptors (174,175). Interestingly, treatment of HepG2 cells in vitro with TSH stimulated PCSK9 expression and decreased LDL receptors (175)

 

In addition to the effects on the LDL receptor levels, other changes induced by thyroid hormone may also contribute to the increases in LDL-C levels in hypothyroid patients. Studies in LDL receptor deficient mice (LDL receptor knock-out mice) have shown that thyroid hormone administration lowers LDL-C levels despite the absence of LDL receptors(176,177).Thyroid hormone also stimulates the conversion of cholesterol to bile acids by increasing cholesterol 7 alpha hydroxylase, the initial enzyme in bile acid synthesis, and in hypothyroid patients a decrease in bile acid synthesis could contribute to an increase in LDL-C levels (174,177-180). Furthermore, the expression of ABCG5 and ABCG8, the transporters that mediate the movement of cholesterol from the hepatocyte into the bile, are also stimulated by thyroid hormone (181,182). In addition, studies by Goldberg and colleagues demonstrated that thyroid hormone decreases Apo B production and hence in hypothyroidism there could be an increase in Apo B synthesis (176). Finally, studies have shown that hypothyroidism is associated with increased intestinal cholesterol absorption that is due to an increase in NPC1L1 (181). Thus, a number of potential pathways could contribute to the increased LDL-C that occurs in hypothyroidism.

 

TRIGLYCERIDES

 

As noted above hypothyroidism has only modest effects on plasma triglyceride levels. Several but not all studies have shown that thyroid hormone stimulates lipoprotein lipase activity (183-188). A decrease in lipoprotein lipase activity could lead to the decreased clearance of triglyceride rich lipoproteins accounting for the increase in plasma triglyceride levels in hypothyroidism. Moreover, studies have shown that thyroid hormone stimulates the expression of Apo A-V, which potentiates the activity of lipoprotein lipase, and is associated with decreases in plasma triglyceride levels (189). Additionally, thyroid hormone decreases angiopoietin-like proteins 3 and 8, inhibitors of lipoprotein lipase, and the levels of angiopoietin-like proteins 3 and 8 are elevated in hypothyroid patients which could lead to a decrease in lipoprotein lipase activity (190,191). Lastly, hypothyroidism increases hepatic VLDL-TG secretion rate, which could also contribute to elevations in plasma triglyceride levels (192).

 

HDL-C

 

As noted above hypothyroidism has only modest effects on plasma HDL-C levels. However, thyroid hormone might be having effects on HDL metabolism that are not reflected in HDL-C levels, as a number of key proteins involved in HDL metabolism and reverse cholesterol transport are regulated by thyroid hormone. Specifically, CETP, hepatic lipase, LCAT, and SR-B1 are increased by thyroid hormone and are decreased in hypothyroidism (182,183,185,188,193-200). A decrease in CETP, hepatic lipase, LCAT, and SR-B1 would be anticipated to result in a decrease in reverse cholesterol transport (115). Moreover, sera from animals treated with thyroid hormone have the increased ability to facilitate the efflux of cholesterol from macrophages to HDL via ABCA1 (201).

 

LIPOPROTEIN (a)

 

The mechanism for the increase in Lp(a) is unknown.

 

HYPERTHYROIDISM

 

Effect of Hyperthyroidism on Lipid and Lipoprotein Levels

 

In hyperthyroidism total cholesterol and LDL-C levels are decreased (132,202). Additionally, HDL-C and Lp(a) levels are also decreased (132,202) (Table 6). The effect on triglyceride levels is variable and triglyceride levels may be increased, decreased, or unchanged (132,202). Restoration of euthyroidism results in the normalization of lipid and lipoprotein levels. A meta-analysis reported that treatment of hyperthyroidism resulted in a significant increase in total cholesterol (44.5mg/dL; 95% CI: 38.0 - 51.0), LDL-C (31.1mg/dL; 95% CI 24.3- 37.9), HDL-C (5.52mg/dL; 95% CI 1.48- 9.56), Apo AI (15.6 mg/dL; 95% CI; 10.4- 20.8), apo B (26.1mg/dL; 95% CI 22.7- 29.6), and Lp[a] (4.18mg/dL; 95% CI; 1.65- 6.71) with no significant change in triglyceride levels (146). Treatment of subclinical hyperthyroidism did not change any lipid parameters significantly (146). A recent small study reported that patients with severe subclinical hyperthyroidism (TSH <0.1 mlU/L) treated with radioactive iodine had increases in total cholesterol (16.7 ± 4.5mg/dL; p < 0.01), LDL-C (14.3 ± 4.1mg/dL; p < 0.01) and triglycerides (25.2±9.4mg/dL; p< 0.01) while patients with mild subclinical hyperthyroidism (TSH: 0.1-0.39 mlU/L) did not demonstrate statistically significant increases in lipid levels (203).

 

Table 6. Effect of Hyperthyroidism on Lipid and Lipoprotein Levels

 

Total Cholesterol

Decrease

LDL-C

Decrease

HDL-C

Decrease

Triglycerides

Variable

Lp(a)

Decrease

Apo B

Decrease

Apo A-I

Decrease

 

Given the beneficial effects of thyroid hormone on lipid and lipoprotein levels, consideration has been given to treating patients with thyroid hormone/thyroid hormone analogues to reduce cardiovascular disease. The Coronary Drug Project examined the use of D-thyroxine for lipid lowering in patients with cardiovascular disease. While D-thyroxine was effective in lowering LDL-C levels, it was also associated with an increase in cardiovascular deaths and the trial was therefore stopped early (204). More recently there have been efforts by the pharmaceutical industry to develop thyroid hormone analogs and mimetics that would have the beneficial effects of thyroid hormone on lipids and lipoproteins without inducing the harmful effects of excess thyroid hormone (205).

 

Mechanism for the Changes in Lipids and Lipoproteins in Hyperthyroidism

 

Thyroid hormone regulates the expression and activity of a number of key enzymes and receptors that regulate lipid and lipoprotein levels. For details see section on hypothyroidism.

 

LDL-C

 

The decrease in LDL-C levels is primarily due to an increase in hepatic LDL receptors resulting in the accelerated clearance of circulating LDL (132). This increase in LDL receptors is due to thyroid hormone stimulating the increased expression of LDL receptors (132,172,173). In addition, hyperthyroidism leads to a decrease in PCSK9, which will lead to a decrease in the degradation in LDL receptors contributing to the increase in LDL receptors (174). 

 

Studies in LDL receptor deficient mice (LDL receptor knock-out mice) have shown that thyroid hormone administration lowers LDL levels despite the absence of LDL receptors, indicating that factors in addition to up-regulation of the LDL could contribute to the decrease in circulating LDL [167, 168]. Thyroid hormone stimulates the elimination of cholesterol from the body by increasing the conversion of cholesterol into bile acids and increasing the biliary secretion of bile acids and cholesterol (174,179,180,206). Thyroid hormone also diminishes intestinal absorption of dietary cholesterol (181). Finally, thyroid hormone decreases Apo B production and hence hyperthyroidism could result in a decrease in Apo B synthesis [167]. The relative contribution of these changes in contributing to the decrease in LDL-C is unknown.

 

HDL-C

 

A number of key proteins involved in HDL metabolism and reverse cholesterol transport are regulated by thyroid hormone. Specifically, CETP, hepatic lipase, LCAT, and SR-B1 are increased by thyroid hormone (182,183,185,188,193-200). An increase in CETP, hepatic lipase, LCAT, and SR-B1 would be anticipated to result in a decrease in HDL-C and an increase in reverse cholesterol transport (115). Moreover, sera from animals treated with thyroid hormone have the increased ability to facilitate the efflux of cholesterol from macrophages to HDL via ABCA1 (201).

 

LIPOPROTEIN (a)

 

The mechanism for the decrease in Lp(a) is unknown. Studies have shown that decreases in PCSK9 activity can reduce Lp(a) levels so perhaps the thyroid hormone induced decrease in PCSK9 plays a role (174,207).

 

CUSHING’S SYNDROME

 

Effect of Cushing’s Syndrome on Lipid and Lipoprotein Levels

 

It is difficult to state the true prevalence of hyperlipidemia in patients with Cushing’s syndrome due to the fact that cut-offs used to establish the presence of hyperlipidemia vary among different studies and the number of patients in these studies have been relatively small. Additionally, the severity of the Cushing’s syndrome is also a key variable. Nevertheless, it is apparent that dyslipidemia is a common feature of Cushing’s syndrome with an elevation in plasma triglycerides and total cholesterol due to an increase in circulating VLDL and LDL (208-214). The elevation in total and LDL-C levels correlates with the severity of the Cushing’s syndrome (208,210). A comparison of ACTH-dependent and ACTH-independent Cushing syndrome did not observe differences in lipid levels (215). The central obesity that characterizes Cushing’s syndrome likely contributes to the dyslipidemia with patients who have central obesity more likely to have alterations in lipid levels (214). Additionally, if Cushing’s syndrome is associated with diabetes this can further alter lipid and lipoprotein levels (216). These alterations in lipid and lipoprotein levels improve or normalize after treatment and lowering of the elevated cortisol levels (208,217). The effect of Cushing’s syndrome on HDL-C is more variable with increases and decreases in HDL-C both being reported in different studies (208,209). Finally in one small study Lp(a) levels were not altered in patients with Cushing’s syndrome (218), while in another small study Lp(a) levels were increased (214).

 

Most series report improvement in hyperlipidemia with correction of elevated cortisol levels, though a complete normalization of lipid parameters is frequently not achieved (208). In a longitudinal study, 25 patients had a significant decrease in LDL-C levels after one year of normalization of cortisol levels, but levels still remained higher than healthy controls, albeit similar to BMI-matched controls (213). Similarly, in a cross-sectional study carried out 5 years after cure or control of pituitary Cushing’s disease, levels of total and LDL-C were similar to levels found in BMI-matched controls, but higher than in normal controls (214). 

 

Table 7. Effect of Cushing’s Syndrome on Lipid and Lipoprotein Levels

Total Cholesterol

Increase

LDL-C

Increase

HDL-C

Variable

Triglycerides

Increase

Lp (a)

No change or increase

Apo B

Increase

Apo A-I

Variable

 

In patients without inflammatory disorders, the administration of glucocorticoids has variable effects on the lipid profile; HDL-C levels are typically increased with the magnitude of change in plasma triglyceride and LDL-C varying among studies (219-221). In patients with inflammatory diseases, the effect of glucocorticoids on lipids is confounded by the marked anti-inflammatory effects of glucocorticoids. Inflammation affects lipid and lipoprotein levels and thus reducing inflammation per se can affect the lipid response to glucocorticoid treatment (222). Similarly, the effect of glucocorticoids on lipids following transplantation or the treatment of other medical conditions is also difficult to interpret due to the simultaneous use of other medications and the response of the underlying medical conditions. Furthermore, the dose and route of administration of the glucocorticoids can be an important variable, as low doses often have minimal effects on triglyceride, LDL-C, and HDL-C levels while high doses tend to increase serum triglyceride, LDL-C, and HDL-C levels.

 

Table 8. Effect of Glucocorticoid Treatment on Lipid and Lipoprotein Levels

Total Cholesterol

Increase

LDL-C

No Change or Increase

HDL-C

Increase

Triglycerides

No Change or Increase

 

Mechanism for the Changes in Lipids and Lipoproteins in Cushing’s Syndrome

 

The mechanisms by which excess glucocorticoids induce changes in lipid and lipoprotein metabolism have not been precisely elucidated and the literature on this topic is often contradictory (223,224). Below we will review some of the potential mechanisms that could account for the observed changes.

 

LDL-C

 

A single study in rats has shown that glucocorticoids decrease hepatic LDL receptor expression (225). However, this glucocorticoid effect on LDL receptor expression was not seen by Galman and colleagues (181). Intriguingly, Galman and colleagues reported that ACTH stimulation of the adrenals did decrease the expression of both hepatic LDL receptors and SR-B1 receptors, suggesting that hormones other than glucocorticoids might have effects on liver receptors (226). Whether this plays a role in the increase in plasma LDL-C levels seen in some individuals with Cushing’s syndrome is unknown.

 

TRIGLYCERIDES

 

Glucocorticoid administration stimulates hepatic fatty acid synthesis by increasing the activity of acetyl CoA carboxylase and fatty acid synthesis (223,227-230). In addition, glucocorticoids also stimulate the enzymes required for the synthesis of triglyceride in the liver (231-233). The increase in hepatic triglyceride levels leads to the decreased degradation of Apo B and an increase in the formation and secretion of VLDL (223,224,230,234-236). Moreover, in patients with Cushing’s syndrome VLDL production rates are increased, while VLDL clearance is not altered, indicating that hepatic overproduction of VLDL accounts for the increase in serum triglyceride levels (217). This increase in VLDL production could also contribute to the increase in LDL-C levels in patients with Cushing’s syndrome (217).

 

In addition to glucocorticoids increasing hepatic fatty acid synthesis, in acute experimental models, glucocorticoids also increase adipose tissue lipolysis resulting in an increase in circulating free fatty acid levels (223,224,237-243). Glucocorticoids increase the expression of adipose tissue triglyceride lipase and hormone sensitive lipase, two of the key enzymes that mediate the breakdown of triglycerides into free fatty acids in adipose tissue (238,241,244,245). Furthermore, glucocorticoids also stimulate adipose tissue lipolysis by increasing cAMP levels, which stimulates the activation of protein kinase A (PKA) leading to the phosphorylation of hormone sensitive lipase and perilipin (237,241). However, studies have shown that chronic elevations in glucocorticoids do not increase adipose tissue lipolysis; thus it is not clear whether increased transport of fatty acids from adipose tissue to liver contributes to the chronic increased formation and secretion of VLDL by the liver (224,246,247

 

HDL-C

 

Studies have shown that glucocorticoids increase the synthesis and secretion of Apo A-I by direct effects on the Apo A-I promoter that are mediated via the glucocorticoid receptor (248,249). The increased production of Apo A-I could lead to an increase in HDL-C. Furthermore, glucocorticoids decrease hepatic lipase activity and increase LCAT activity, which could also contribute to an increase in HDL-C levels (250).

 

Risk of Cardiovascular Disease

 

Patients with Cushing’s syndrome have a higher mortality rate than age and gender matched controls, which is mainly due to an increased risk of cardiovascular disease (251-254). Notably this increased mortality risk remains even after remission of Cushing’s syndrome, but is reduced compared to persistent disease (254,255). Furthermore, studies have shown that the hazard ratio for myocardial infarctions was 3.7 and for strokes was 2.0 in patients with Cushing’s syndrome (256). Moreover, patients with Cushing’s syndrome have an increase in carotid intima-media thickness, which persists after remission of the disease (213,214,257-259). Additionally, coronary artery calcium, a marker of atherosclerosis, is also increased in patients with Cushing’s syndrome and also persists after disease remission (260,261). Importantly, iatrogenic Cushing’s syndrome also increases the risk for cardiovascular disease (262-265). Thus, it is quite clear that Cushing’s syndrome increases the risk and occurrence of atherosclerotic cardiovascular disease. It is likely that the dyslipidemia that accompanies Cushing’s syndrome contributes to the increase in atherosclerotic cardiovascular disease, but it must be recognized that Cushing’s syndrome also induces other abnormalities that are highly associated with an increased risk of atherosclerotic cardiovascular disease such as central obesity, diabetes, insulin resistance, hypercoagulability, and hypertension (266,267). It is therefore likely that the increase in atherosclerotic cardiovascular disease seen in patients with Cushing’s syndrome is multifactorial.

 

Because of an increased risk of cardiovascular disease the Endocrine Society recommends that “in adults with persistent endogenous Cushing syndrome, we suggest statin therapy, as adjunct to lifestyle modification, to reduce CV risk irrespective of the CV risk score” with a goal LDL-C < 70mg/dL (268). Additionally, the Endocrine Society recommends “In adults with cured Cushing syndrome, we advise the approach to CV risk assessment and treatment be the same as in the general population” (268).

 

Effect of Drugs Used to Treat Cushing’s Syndrome on Lipid Levels

 

Ketoconazole is used to treat patients with Cushing’s syndrome.  It is an anti-fungal imidazole derivative that blocks several steps in cortisol biosynthesis thereby lowering serum cortisol levels.  However, ketoconazole is also an inhibitor of cholesterol biosynthesis, acting directly by blocking the conversion of methyl sterols to cholesterol and indirectly by suppressing cholesterol synthesis via feedback inhibition of HMG-CoA reductase by sterol intermediates (269,270).  In the past, ketoconazole had been used to treat patients with familial hypercholesterolemia before the widespread use of statins, as it reduced total, intermediate density cholesterol, LDL-C, and apo B levels by approximately 25% (271).  Thus, its use to control hypercortisolism may have a beneficial effect on lipid and lipoprotein levels. Levoketoconazole, also decreases total cholesterol and LDL-C levels by approximately 25% and slightly increases HDL-C levels (272).

 

It is important to recognize that ketoconazole and levoketoconazole also interfere with the metabolism of many drugs through the inhibition of several hepatic P450 enzymes.  Simvastatin, lovastatin, and atorvastatin are all metabolized by cytochrome P450 CYP3A4, and thus, their plasma concentrations and risk of myotoxicity are greatly increased with concomitant ketoconazole therapy (273).  Pravastatin, pitavastatin, and rosuvastatin are preferable as their plasma concentrations are not significantly increased by CYP3A4 inhibitors (273).

 

Mitotane is used for treatment of adrenal carcinoma or intractable Cushing’s disease and results in adrenocortical atrophy and necrosis and inhibits steroidogenesis.  Mitotane raises circulating cholesterol, LDL-C, Apo B, and HDL-C levels (274-276).  Changes in triglyceride levels are variable (276). In one report no changes Lp(a) levels were observed (274).  Mitotane increases HMGCoA reductase activity, which may contribute to the increase in LDL-C (277). The increase in LDL-C levels has been shown to be decreased by treatment with statins (274,276). Because mitotane induces CYP3A4 activity one should use a statin that is not metabolized by this enzyme (for example pravastatin or rosuvastatin) (276). In a case report mitotane increased LDL-C levels as high as 300mg/dl (278).

 

Mifepristone, a potent antagonist of glucocorticoid and progesterone receptors, lowers HDL-C and Apo AI levels (279). The mechanism for this decrease in HDL-C is unknown. In a small study short-term administration of mifepristone reduced serum triglyceride levels, which correlated with increases in adipose tissue lipoprotein lipase activity (280).

 

Pasireotide is a somatostatin analogue and in patients with Cushing’s syndrome has been shown to decrease total cholesterol and LDL-C levels (281-283). In some studies triglyceride levels were also decreased (282).

 

TESTOSTERONE

 

Effect of Testosterone on Lipid and Lipoprotein Levels

 

ENDOGENOUS TESTOSTERONE LEVELS

 

Numerous observational (epidemiological) studies have shown that serum testosterone levels directly correlate with HDL-C and Apo A-I levels (i.e. subjects with low serum testosterone levels have lower HDL-C and Apo A-I levels) (284-290). Moreover, low serum testosterone levels are inversely correlated with total cholesterol, LDL-C, Apo B, and triglyceride levels (i.e. subjects with low testosterone levels have higher total cholesterol, LDL-C, Apo B, and triglycerides) (285,289-291). Thus, individuals with low serum testosterone levels have a pro-atherogenic lipoprotein pattern with low HDL-C levels and high triglyceride and LDL-C levels.

 

Not unexpectedly, given the low HDL-C levels and high triglyceride levels, individuals with low serum testosterone levels are more likely to have the metabolic syndrome (290,292,293). It should be recognized that these associations do not necessarily imply that the low serum testosterone levels are causative. For example, it is likely that obesity and related metabolic abnormalities, such as type 2 diabetes, lead to both the abnormal lipid pattern and the low serum testosterone levels. Indeed, obesity is associated with low testosterone and weight loss restores testosterone levels (293-297). Thus, observational studies may be confounded.

 

Table 9. Correlation of Testosterone Levels with Lipid and Lipoprotein Levels

HDL-C

Positive (low T = lower)

LDL-C

Negative (low T = higher)

Triglycerides

Negative (low T = higher)

Non-HDL-C

Negative (low T = higher)

Lp(a)

Negative (low T = higher)

 

ANDROGEN DEPRIVATION THERAPY

 

Studies of the effect of androgen deprivation therapy not only constitute a clinically relevant state but also provide an alternative approach to understanding the effects of low testosterone levels on lipid and lipoprotein levels. In contrast to the associations in observational studies, most studies of androgen deprivation therapy have shown an increase in plasma HDL-C and Apo A-I levels (298-305). This increase occurs very rapidly within 2 weeks of lowering serum testosterone levels (298). Furthermore, this increase in HDL-C is inhibited if one simultaneously administers testosterone demonstrating that this increase is due to the suppression of testosterone levels (303). In addition, androgen deprivation therapy is also associated with an elevation of LDL-C, non-HDL-C, Lp(a), and triglyceride levels (299-302,304,306-309). The increase of Lp(a) is notable as the metabolism of Lp(a) often does not parallel the metabolism of LDL.

 

Table 10. Effect of Androgen Deprivation Therapy on Lipid and Lipoprotein Levels

HDL-C

Increase

LDL-C

Increase

Triglycerides

Increase

Non-HDL-C

Increase

Lp(a)

Increase

 

TESTOSTERONE TREATMENT

 

There have been several meta-analyses that have examined the effect of testosterone treatment on lipid and lipoprotein levels but the results have been variable. Baseline differences, type of therapy, and duration of therapy may contribute to the differing results. A meta-analysis by Whitsel and colleagues demonstrated that total cholesterol, LDL-C, and HDL-C levels decreased after intramuscular  testosterone treatment, but triglyceride levels did not change (310).  A meta-analysis by Isidori also demonstrated a decrease in HDL-C levels, but found no change in LDL-C with testosterone treatment (both intramuscular and transcutaneous) (311) . Similarly, a meta-analysis by Fernández-Balsells and colleagues demonstrated a decrease in HDL-C levels but no change in LDL-C or triglyceride levels with testosterone treatment (both intramuscular and transcutaneous administration) (312). A meta-analysis by Haddad et al failed to demonstrate any significant changes in HDL-C, LDL-C, or triglyceride levels (313). A recent meta-analysis by Corona and colleagues did not find changes in HDL-C levels but reported small decreases in total cholesterol and triglycerides (314). A meta-analysis of testosterone replacement therapy in patients with type 2 diabetes found a decrease in triglycerides, total cholesterol, and LDL-C and an increase in HDL-C (315). This improvement in lipid parameters could be due to decreases in body weight, glycemic control, and insulin resistance. Finally, a non-randomized long term trial (8 years) of intramuscular testosterone therapy in patients with pre-diabetes resulted in a decrease in body weight and a decrease in A1c that was accompanied by decreases in LDL and non-HDL-C and triglyceride levels and increases in HDL-C levels compared to an untreated comparison group suggesting that long-term therapy might be beneficial on lipids by effecting body weight and glucose homeostasis (316).

 

The reason for the differences between these meta-analyses is likely due to the fact that the changes in lipid and lipoprotein levels induced by testosterone treatment are relatively small and variable depending upon the patient population studied, the route and dose of testosterone administration, the duration of therapy, the specific testosterone preparation (whether or not it can undergo aromatization to estrogens), and perhaps other unrecognized factors. For example, the reductions in HDL-C appear to be greater in patients whose baseline HDL-C levels are high (311,312). Additionally, transdermal testosterone treatment appears to have less effect on HDL-C levels than intramuscular administration (317). High dose testosterone treatment appears to more consistently lower HDL-C levels than does low dose treatment (318). For example, testosterone enanthate 200mg IM every week used in a contraception study resulted in a relatively robust 13% decrease in HDL-C levels (319). Similarly, raising serum testosterone levels to higher levels produces greater decreases in LDL-C levels (318). Finally, using testosterone preparations that are not converted to estrogens or simultaneously blocking aromatization can lead to more profound decreases in HDL-C and LDL-C levels, which can be attributed to estrogens having effects on lipid and lipoprotein levels that counterbalance the effects of androgens (estrogens increase HDL-C and decrease LDL-C) (320,321). The important clinical point is that in the typical androgen deficient patients that we treat with the usual testosterone therapy there will only be a modest or no changes in plasma lipid and lipoprotein levels. The minimal effect of testosterone therapy was clearly demonstrated in a large randomized double-blind trial of 788 males over the age of 65 with low testosterone levels who were treated with either testosterone gel to normalize testosterone levels or placebo for 1 year (322). In this trial HDL-C (adjusted mean difference, -2.0 mg/dL; P < 0.001), and LDL-C were both slightly decreased (adjusted mean difference, -2.3 mg/dL; P = 0.051) from baseline with no change in triglyceride levels in the testosterone treated individuals.

 

While treatment of typical older hypogonadal men with testosterone therapy has only modest to no effects on plasma lipids and lipoproteins, the use of high dose androgenic steroids in young men for the purpose of increasing muscle mass and strength can have profound effects. In a study by Webb and colleagues of 14 individuals taking high dose androgenic  steroids, HDL-C levels were markedly reduced to 29mg/dl, which was less than 50% of the mean HDL-C when exogenous steroids were not used (61mg/dl) (323). Additionally in these individuals LDL-C levels were also higher on androgenic steroids (150mg/dl) than off of androgenic steroids (125mg/dl) (323). Similarly, Hurley and colleagues demonstrated that androgen use by eight bodybuilders and four powerlifters lowered HDL-C levels by 55% and raised LDL-C levels by 61% (324). In a double blind cross-over study anabolic steroids, which may not have androgenic effects, induced a 25-27% decrease in HDL-C levels, which returned towards normal 6 weeks after cessation of drug use (325). Thus, if one sees an athletic male with unexpectedly low HDL-C levels one should suspect androgen and/or anabolic steroid use, which is often obtained as a dietary supplement or as a pharmaceutical from an unregulated source.

 

There are a number of potential explanations why the changes in lipid and lipoprotein levels are greater in athletes using androgenic steroids. First, the doses used by the athletes are much higher than used in typical testosterone replacement. Second, the androgenic steroids used are often different and more potent (for example nandrolone-decanoate and oxandrolone). Often the compounds used are not converted to estrogen by aromatase and therefore their effects on serum lipid levels will not be counterbalanced by estrogen formation [265, 272]. Third, aromatase inhibitors are sometimes used simultaneously in combination with the androgenic steroids. Lastly, young athletes are often lean and have little adipose tissue and thus low aromatase activity. There can be individual patient variation in aromatase activity with obese older individuals having increased aromatase activity compared to young athletic individuals (326). As noted earlier, the conversion of testosterone to estrogens by aromatase may blunt the effects of testosterone as estrogens will increase HDL-C levels and decrease LDL-C levels. Together it is likely that these factors account for the more robust changes in lipids and lipoprotein levels induced by androgens in young athletes.

 

TRANSGENDER MALES

 

Testosterone therapy in transmen results in an increase in LDL-C levels and a decrease in HDL-C levels with some studies also showing an increase in triglyceride levels (327-331). In transmale adolescents treated with testosterone LDL-C levels increased and HDL-C levels decreased compared to cisgender females (332). These changes are likely due to the combination of an increase in testosterone and a decrease in estrogen.

 

LIPOPROTEIN (a)

 

There is a trend towards a higher incidence of clinically significant elevations in Lp(a) levels in men with low testosterone levels (333). Additionally, reductions in serum testosterone levels by orchiectomy or treatment with GnRH antagonists results in an increase in Lp(a) levels (305,334). Conversely, several studies have shown that testosterone administration decreases Lp(a) levels and the effect is more robust in individuals who have high baseline Lp(a) levels (319,335,336). Moreover, it has been shown that simultaneously administering testosterone with an aromatase inhibitor does not markedly reduce the ability of testosterone to decrease Lp(a) levels, indicating that the conversion of testosterone to estrogens does not account for this effect suggesting a direct action of testosterone (335). Lp(a) is a pro-atherogenic lipoprotein so testosterone induced decreases should be beneficial.

 

SUMMARY

 

The most consistent effects of androgen therapy on lipid and lipoprotein levels are to decrease HDL-C and Lp(a) levels. These effects are most apparent with high dose testosterone therapy. The decreases in HDL-C and Lp(a) levels with testosterone therapy are consistent with the increases seen with androgen deprivation therapy. However, both types of treatment result in changes that are the opposite of those seen in the observational studies, suggesting that the observational studies are confounded. However, high potency androgen therapy in young healthy men tends to increase LDL-C levels and markedly decrease HDL-C levels (337).

 

Table 11. Effect of Testosterone Therapy on Lipid and Lipoprotein Levels

HDL-C

Decreased or No Change

LDL-C

Decrease

Triglycerides

No consistent change

Lp(a)

Decrease

 

Mechanism for the Testosterone Induced Lipid and Lipoprotein Changes

 

HDL-C

 

The decrease in HDL-C levels with testosterone administration has been attributed to increases in the expression of SR-B1 in the liver and increases in plasma hepatic lipase activity. In Hep G2 cells, the addition of testosterone increased the mRNA and protein levels of SR-B1 and hepatic lipase but had no effect on the expression of Apo A-I or ABCA1 (338). Moreover, androgen administration increased plasma hepatic lipase activity but had little effect on lipoprotein lipase (320,339-342). An increase in SR-B1 in the liver will facilitate the transfer of cholesterol from HDL particles into the hepatocyte, decreasing plasma HDL-C levels (115). An increase in hepatic lipase activity will increase the hydrolysis of triglycerides and phospholipase on HDL, resulting in the formation of smaller HDL particles, the release of Apo A-I, and increased Apo A-I degradation leading to a decrease in plasma HDL levels (115). Thus, the increase in SR-B1 and hepatic lipase induced by androgens could account for the decrease in HDL-C levels seen with testosterone treatment. There is the potential that the increase in SR-B1 is protective in atherosclerosis as it enhances reverse cholesterol transport from HDL (115).

 

LDL-C

 

The mechanism by which testosterone therapy might affect LDL-C levels is uncertain. It has been shown that testosterone can antagonize the ability of estrogens to stimulate LDL receptor expression in the liver, which could lead to a decrease in hepatic LDL receptors and an increase in plasma LDL-C levels (343).

 

LIPOPROTEIN (a)

 

The mechanism by which testosterone treatment lowers Lp(a) levels is unknown.

 

Risk of Cardiovascular Disease

 

In the Endocrinology of Male Reproduction section of Endotext the chapter by Yeap and Dwivedi (“Androgens and Cardiovascular Disease in Men”), extensively reviews the literature on the linkage of testosterone and cardiovascular disease (344). Therefore, we will only briefly summarize the relevant information.

 

ENDOGENOUS TESTOSTERONE LEVELS

 

There have been numerous cross-sectional studies of testosterone levels in patients with coronary artery disease vs. controls and the results have varied (344). Some studies have shown no association while other studies have found low testosterone levels in patients with coronary artery disease. The majority of prospective studies have shown that cardiovascular disease occurs more frequently in subjects with low testosterone levels. Whether the low testosterone is causative or a biomarker of poor cardiovascular health (e.g., obesity, metabolic syndrome, diabetes) cannot be determined from these types of observational studies.

 

ANDROGEN DEPRIVATION THERAPY

 

In a meta-analysis by Zhao and colleagues of population-based observational studies comparing androgen deprivation therapy in patients with prostate cancer vs. controls with prostate cancer, six studies were identified with a total of 129,802 androgen deprivation therapy patients and 165,605 controls (345). In this analysis, cardiovascular disease was increased by 10% and cardiovascular mortality by 17% in the androgen deprivation therapy patients. In a meta-analysis by Carneiro and colleagues of 126,898 prostate cancer patients in four cohort studies and 10,760 prostate cancer patients in nine randomized controlled trials, these authors found that cardiovascular events were increased two fold in the androgen deprivation groups (346). When only the randomized trials were analyzed, the relative risk was increased 1.55-fold in the androgen deprivation patients. In contrast, a meta-analysis by Nguyen and colleagues of 8 randomized trials with 4141 patients did not find an increased risk of cardiovascular disease (347). Finally, a meta-analysis by Bosco of eight observational studies reported a relative risk of 1.57 for fatal and non-fatal cardiovascular disease in patients with prostate cancer treated with GnRH agonists (348). These and other results suggest that the risk of cardiovascular disease is increased in men undergoing androgen deprivation therapy, despite the increase in HDL-C.

 

TESTOSTERONE TREATMENT

 

There have been a large number of observational studies of the risk of cardiovascular disease in men treated with testosterone replacement and the results have been inconsistent, with some studies showing that testosterone increases the risk while other studies have shown no increase in risk (344). Interestingly, in a very large retrospective study of 544,115 testosterone treated patients it was reported that men treated with intramuscular testosterone had an increased risk of cardiovascular events (1.26) and death (1.34), whereas individuals treated with either testosterone gel or patch did not have an increased risk (349).

 

With regards to randomized trials, the Testosterone in Older Men with Mobility Limitations Trial (TOM trial) reported an increase in cardiovascular events with testosterone treatment (350). This trial studied 209 men with an average age of 74 years who had a high baseline prevalence of cardiovascular disease (53%) and major cardiovascular risk factors (diabetes 24%, hypertension 85%, and hyperlipidemia 63%). In this trail subjects were treated with high doses of testosterone gel that resulted in high serum testosterone levels.  Although 23 subjects in the testosterone group and 5 in the placebo group had a cardiovascular-related adverse event, it should be recognized that many of these cardiovascular events were not atherosclerotic; only 7 men in the testosterone group and 1 in the placebo group had an atherosclerosis related event. Of note, a similar trial using lower doses of testosterone did not observe an increase in cardiovascular events (351). Additionally, a recent randomized trial with 308 men 60 years or older with low or low-normal testosterone levels demonstrated that treatment with testosterone gel for 3 years did not result in a significant difference in the rates of increase in either common carotid artery intima-media thickness or coronary artery calcium (352). In contrast, a randomized trial demonstrated that testosterone treatment compared with placebo was associated with a significantly greater increase in noncalcified plaque volume from baseline to 12 months (from median values of 204 mm3 to 232 mm3 vs 317 mm3 to 325 mm3, respectively; estimated mean difference, 41 mm3; 95% CI, 14 to 67 mm3; P = .003) with no difference in progression of coronary calcium scores (353). It should be noted that baseline plaque volume differed between the testosterone and placebo group, which complicates interpretation of these results.

 

With the exception of one meta-analysis by Xu et al (354), most meta-analyses of randomized clinical trials of testosterone therapy have not demonstrated a statistically significant difference in the occurrence of cardiovascular events (312,313,355-362). Of note, one meta-analysis explored the effect of the route of administration of testosterone and reported that oral testosterone treatment significantly increased cardiovascular risk (RR = 2.20), while neither intramuscular nor transcutaneous delivery (gel or patch) significantly altered cardiovascular risk (355).

 

To definitively determine the effect of testosterone replacement therapy on cardiovascular disease will require a large randomized outcome trial similar to the Women’s Health Initiative. The TRAVERSE study is a large randomized trial designed to definitively answer this crucial question (363)

 

SUMMARY

 

While the data suggests that androgen deprivation therapy increases the risk of atherosclerotic cardiovascular disease, the effect of testosterone administration is unclear.

 

FEMALE SEX STEROID HORMONES

 

Effect of Female Sex Steroid Hormone on Lipid and Lipoprotein Levels

 

PREMENOPAUSAL WOMEN

 

The plasma lipid profile of premenopausal women is less pro-atherogenic than the lipid profile in men (364-367). Specifically, HDL-C levels are increased (approximately 10mg/dl higher in women), while LDL-C and non-HDL-C levels are slightly lower compared to male values (364-367). Additionally plasma triglyceride levels are also decreased and the average size of LDL particles is increased in premenopausal women compared to men (364-367).

 

Notably most of these differences emerge during puberty. Prior to puberty the lipid profiles of girls and boys are very similar but during puberty HDL-C levels in boys decrease while in girls the HDL-C levels do not change (364-367). Additionally, during puberty triglyceride levels increase in boys with no change in triglyceride levels occurring in girls. LDL-C levels are similar in boys and girls before and during puberty but after age 20 LDL-C increase in both males and females but the increase is greater in males resulting in a modest difference in LDL-C levels between the sexes (364-367).

 

Table 12. Comparison of Lipid and Lipoprotein Levels in Premenopausal Women Compared to Men

Lipids/Lipoprotein

Premenopausal Women Compared to Men

LDL-C

Lower

HDL-C

Higher

Triglycerides

Lower

Non-HDL-C

Lower

 

POSTMENOPAUSAL WOMEN

 

The changes in lipids and lipoproteins that occur during menopause are relatively small and therefore the results reported in the literature are variable (364-367). Cross-sectional studies tend to show a greater shift towards a pro-atherogenic lipid profile after the menopause whereas in longitudinal studies the changes are smaller (364-367). In post-menopausal women increases in LDL-C are reported in most, but not all studies, and the composition of LDL shifts towards smaller dense LDL particles (364-367). HDL-C levels tend to be stable but some studies have reported small decreases (364-367). Following surgical menopause the above changes tend to be more rapid and robust and in this setting Lp(a) levels have been reported to increase; however, during natural menopause the change in Lp(a) is very modest (368,369). It is important to recognize that during menopause there are changes in factors in addition to the loss of sex steroid hormones that can alter lipid and lipoprotein levels. Menopause is associated with increases in total and central body fat and a decrease in insulin sensitivity, which are well recognized to affect lipid and lipoprotein metabolism (37).

 

Table 13. Effects of Menopause on Lipid and Lipoproteins

Lipids/Lipoproteins

Postmenopausal vs Premenopausal

LDL-C

Increase

HDL-C

No change or small decrease

Lp(a)

No change or increase

 

TRANSGENDER FEMALES

 

In a systemic review and meta-analysis it was reported that in male-to-female individuals, serum TG levels were increased without changes in LDL or HDL-C levels (329). A large observational study of 170 trans females reported an increase in both triglycerides and HDL-C levels (330) but another study only reported an increase in HDL-C levels (331). Additional studies measuring changes in lipid levels in transgender females controlling for estrogen dose, preparation and route of administration, use of other gender affirming therapies, and adjusting for baseline lipid levels are required to better define the changes in lipids that occur.

 

ESTROGEN TREATMENT

 

The effects of oral estrogen treatment on lipids and lipoproteins have been recognized for many years (364,366,370,371). Estrogen administration increases HDL-C levels by 5-15% and decreases LDL-C levels by 5-20% (364,366,370,371). In addition, estrogens also increase triglycerides but in patients with genetic or acquired abnormalities in triglyceride metabolism estrogen therapy can precipitate marked hypertriglyceridemia and even the chylomicronemia syndrome (372). In women with normal baseline triglycerides an approximate 10-15mg/dl increase in triglycerides occurs with estrogen therapy (364,366,370,371). If the increase in triglycerides is substantial, it leads to a decrease in LDL size (i.e., formation of small dense LDL). Not unexpectedly, estrogens induce an increase in Apo A-I levels and a decrease in Apo B levels. Lp(a) levels are also decreased by 20-25% by estrogen therapy (364,366,370,371). The effects of oral estradiol are similar to that of oral conjugated equine estrogens (Premarin).

 

Table 14. Effect of Oral Estrogen Treatment on Lipid and Lipoproteins

Lipids/Lipoproteins

Estrogen Treatment

LDL-C

Decrease

HDL-C

Increase

Triglycerides

Increase

Lp(a)

Decrease

 

Transdermal estrogen administration has less of an effect on lipid and lipoproteins (364,366,370,371,373). The increase in HDL-C and the decrease in LDL-C are markedly blunted (364,366,370,371,373). Importantly, the effect of transdermal estrogen on triglycerides is minimal and therefore in patients with baseline abnormalities in triglyceride metabolism, the use of transdermal estrogen therapy is preferred (364,366,370,371,373). In some studies, treatment with transdermal estradiol has actually decreased plasma triglyceride levels (374). The lack of a robust effect on lipids with transdermal estrogen preparations is likely due to decreased exposure of the liver to estrogens compared with oral therapy. 

 

ESTROGEN AND PROGESTERONE TREATMENT

 

Progestins generally have androgen like effects on lipid and lipoproteins and therefore progestin administration decreases HDL-C and triglyceride levels but has little or no effect on LDL-C levels (364,366,370,371). Thus, when combined with estrogen therapy, the estrogen/progesterone preparation blunts the characteristic estrogen induced increase in HDL-C levels without affecting the estrogen induced reduction in LDL-C levels (364,366,370,371). In many but not all studies, progesterone also blunts the estrogen induced increase in triglyceride levels (364,366,370,371,375). In contrast, progesterone appears to either slightly augment or have no effect on the ability of estrogens to decrease Lp(a) levels (370). It is important to note that the effect of adding progesterone is dependent on both the dose and the androgenicity of the particular progesterone used. Godsland analyzed a large number of studies and found in order of least to most potent progesterone affecting lipid levels the following; dydrogesterone and medrogestone, progesterone, cyproterone acetate, medroxyprogesterone acetate, transdermal norethindrone acetate, norgestrel, and oral norethindrone acetate (370).

 

The Postmenopausal Estrogen/Progestin Intervention (PEPI) trial randomly assigned 875 healthy postmenopausal women to 1) placebo; (2) conjugated equine estrogen (CEE), 0.625 mg/d; (3) CEE, 0.625 mg/d plus cyclic medroxyprogesterone acetate (MPA), 10 mg/d for 12 days/month; (4) CEE, 0.625 mg/d plus continuous MPA, 2.5 mg/day; or (5) CEE, 0.625 mg/d plus cyclic micronized progesterone (MP), 200 mg/day for 12 days/month (375). The effects on plasma lipid and lipoproteins are shown in table 15, which demonstrates that the addition of medroxyprogesterone but not progesterone blunts the estrogen induced increase in HDL-C without affecting the decrease in LDL-C levels. In this particular study medroxyprogesterone did not blunt the estrogen induced increase in triglyceride levels.

 

Table 15. The Effect of Estrogen with or without Progesterone on Plasma Lipid and Lipoprotein Levels (PEPI Trial)

 

Placebo

CEE only

CEE+MPA (cyc)

CEE+MPA (con)

CEE+MP (cyc)

HDL-C

-1.2%

5.6%

1.6%

1.2%

4.1%

LDL-C

-4.1%

-14.5%

-17.7%

-16.5%

-14.8%

Triglycerides

-3.2%

13.7%

12.7%

11.4

13.4%

  

Another study evaluated the effect of hormone replacement on lipid and lipoprotein levels in women with hyperlipidemia (376).  In that study, 58 women with a baseline total cholesterol level of 305mg/dl and LDL-C of 212mg/dl were randomly assigned to treatment with 1.25 mg conjugated estrogen plus medroxyprogesterone acetate 5 mg/day or simvastatin 10 mg daily. The results of this trial are shown in table 16 and demonstrate that statins are more effective in lowering LDL-C levels and have a similar effect on HDL-C as hormone replacement therapy. While statins lower triglyceride levels, hormone replacement therapy increases triglycerides. Of note, hormone replacement therapy markedly lowers Lp(a) levels whereas statin treatment has no effect, on this highly atherogenic particle.  

 

Table 16. Effect of Hormone Replacement Therapy vs. Statin Treatment on Lipid and Lipoprotein Levels

Lipids/Lipoproteins

Hormone Replacement

Simvastatin

Total cholesterol

14% decrease

26% decrease

LDL-C

24% decrease

36% decrease

HDL-C

7% increase

7% increase

Triglycerides

29% increase

14% decrease

Lp(a)

27% decrease

1% increase

 

Considerable variation is seen in the response to hormone replacement therapy. This is likely accounted for by different preparations used, route of administration, dosing regimen (cyclic vs. continuous), difference in hormone status prior to treatment, baseline lipid levels, dietary differences, the presence or absence of other metabolic abnormalities, genetic background, etc. (364,370,371). The studies by Tsuda and colleagues showing that the Apo E phenotype influences the response of LDL to hormonal therapy provide an example of how genetic background can influence response (377). Women with the E2/E2 or E2/E3 genotype demonstrated the largest LDL-C decreases while women with the E4/E4 or E4/E3 genotype had only a small change in LDL-C levels in response to hormonal replacement therapy. Another example of the role of genetics are studies showing that polymorphisms of the estrogen receptor-alpha gene may be associated with an augmented HDL-C rise with estrogen therapy (378).  It is important to note that women who receive cyclic combined therapy (estrogen and progesterone) may have fluctuations in lipoprotein concentrations depending upon the phase of the cycle and it is therefore important to consistently measure lipids during the same hormonal phase, especially when considering starting medications for hyperlipidemia.

 

CONTRACEPTIVES

 

The effect of contraceptives on lipid levels is discussed in the Endotext chapter “Reproductive Health and Its Impact On Lipid Management in Adolescent and Young Adult Females” (379). Table 17 taken from that chapter summarizes the effect of various contraceptives on lipid levels.

 

Table 17. The Effects of Contraceptive Methods on Lipids and Lipoproteins

Contraceptive Method

LDL-C

HDL-C

TG

Comments

Combined Oral Contraceptive Pill

·       Estrogen

Decrease

Increase

Increase

For OCPs with an identical dose of estrogen, the choice and dose of the progestin component may affect net lipid changes

·       Progestin

Increase

Decrease

Decrease

Transdermal Patch

Decrease

Increase

Increase

 

Vaginal Ring

---

---

Increase

 

DMPA

Increase

Decrease

Neutral

 

DMPA = Depot medroxyprogesterone acetate

 

POLYCYSTIC OVARY SYNDROME (PCOS)

 

Women with PCOS characteristically have low HDL-C levels and increased plasma triglyceride levels (380,381). Additionally, LDL-C and non-HDL-C levels are also increased with the LDL being predominantly small dense LDL (380,381). A meta-analysis of 24 studies reported that in women with PCOS triglyceride levels were increased by 26mg/dl, LDL-C by 12mg/dl, non-HDL-C by 19mg/dl and HDL-C was decreased by 6mg/dl (382). The prevalence of elevated Lp(a) levels is also increased in women with PCOS (380,381). It should be noted that the lipid changes in women with PCOS are observed even when the women are not overweight or obese (380,381). In studies of age and weight matched women, the women with PCOS still have lower HDL-C levels and increased triglycerides, LDL-C, and non-HDL-C levels compared to the controls (380,381). The lipid abnormalities in PCOS are likely multifactorial with increases in androgens, decreases in estrogens, obesity, alterations in fat location, insulin resistance, alterations in glucose homeostasis, genetics, and perhaps other factors all contributing to the lipid abnormalities (380,381). Serum PCSK9 concentrations were higher in PCOS patients than normal controls, which could contribute to the increase in LDL-C levels (383). Angiopoietin-like protein 3 levels were increased in PCOS and could contribute to the increase in triglyceride levels (384).  

 

Table 18. Lipid and Lipoprotein Levels in Polycystic Ovarian Syndrome

LDL-C

Increase

HDL-C

Decrease

Triglycerides

Increase

Non-HDL-C

Increase

Lp(a)

Increase

 

Mechanisms for the Female Sex Steroid Induced Lipid and Lipoprotein Changes

    

ESTROGENS

 

There are several effects of estrogen that could lead to an increase in HDL-C levels. First, studies have shown that estrogens stimulate the expression of Apo A-I, which will lead to an increased synthesis of Apo A-I and the increased formation of HDL (366,385-389). Second, estrogen therapy decreases hepatic lipase activity, which will decrease the hydrolysis of triglyceride and phospholipids on HDL particles, which could potentially result in a decrease in the catabolism of HDL (390-392). Finally, estrogens suppress the expression of SR-B1 in the liver, which will decrease the transfer of cholesterol from HDL particles into the hepatocyte increasing plasma HDL-C levels (393). Based on kinetic studies it is likely that the predominant effect of estrogens is to increase the production of HDL, which is mediated by an increase in Apo A-I production (366,385-389). The net result may be protective from atherosclerosis.

 

The decrease in LDL-C induced by estrogen treatment is accounted for by an increase in LDL clearance (366,394-397). Studies have shown that estrogens increase the expression of hepatic LDL receptors (398-401). Additionally, estrogens reduce PCSK9 levels, which would decrease the degradation of LDL receptors (402-404). Together, this would increase the number of hepatic LDL receptors leading to the accelerated clearance of LDL and a reduction in plasma LDL-C levels.

 

The increase in plasma triglyceride levels induced by estrogen treatment is due to the increased production and secretion of VLDL particles (366,388,397,405-407). The mechanism by which estrogens decrease Lp(a) levels is unknown.

 

PROGESTINS

 

Many of the adverse effects of progestins on lipid and lipoproteins, such as decreasing HDL-C levels, are thought to be due to activation of the androgen receptor (i.e. androgenic actions) (408). The considerable variation of progestins in influencing lipid and lipoprotein metabolism are related to their androgenic potency. For detailed information on the effect of testosterone and other androgens on lipid and lipoprotein metabolism see the section above on the mechanism for the testosterone induced lipid and lipoprotein changes.

 

Risk of Cardiovascular Disease

 

PREMENOPAUSAL WOMEN

 

It has been recognized for many years that the risk of cardiovascular disease in premenopausal women is very low and substantially lower than in men of similar age (409-411). There is an approximate 10-year delay in the development of cardiovascular disease in women compared to men. The relative contribution of the less pro-atherogenic lipid profile in women to this sex difference in cardiovascular disease risk is likely important but remains uncertain.

 

POSTMENOPAUSAL WOMEN

 

After the menopause, the risk of cardiovascular disease increases in women (409,410). Of particular note, premature menopause is associated with an increased risk of developing cardiovascular disease, indicating that age is not the sole factor contributing to the increased risk in postmenopausal women (412-415).

 

PREMATURE MENOPAUSE

 

In a meta-analysis of 15 observational studies with 301,438 women it was reported that the risk of cardiovascular disease was higher in women who had premature menopause (age <40 years; HR 1.55, 95% CI 1.38-1.73; p<0·0001), early menopause (age 40-44 years; 1.30, 1.22-1.39; p<0·0001), and relatively early menopause (age 45-49 years; 1.12, 1.07-1.18; p<0·0001) compared to women who had menopause at 50-51 years of age (416). Similarly, a study of 144,260 women using the UK BioBank found that the risk of coronary artery disease was increased in both women with natural premature menopause and surgical premature menopause compared to women with menopause > 40 years of age (surgical premature menopause HR 2.52, p<.001 and natural premature menopause HR 1.39, p< .02). The above studies and others (417-419) indicate the need to evaluate lipids and other cardiovascular disease risk factors in women with premature menopause.        

 

HORMONE REPLACEMENT THERAPY

 

Numerous observational studies have suggested that hormone replacement therapy reduces the risk of cardiovascular disease (420-426). Based on those data, therapeutic trials of hormone replacement therapy were undertaken to see if therapy would prevent or decrease cardiovascular disease. Surprisingly, the randomized clinical trial outcome studies have not demonstrated a uniform decrease in cardiovascular events.

 

HERS Trial

 

The HERS trial was a randomized, blinded, placebo-controlled secondary prevention trial in 2763 women with known coronary artery disease who were postmenopausal with an intact uterus, with a mean age of 66.7 years (427). Patients were randomized to either 0.625 mg of conjugated equine estrogens plus continuous 2.5 mg of medroxyprogesterone acetate or placebo with an average duration of follow-up of 4.1 years. As expected, LDL-C levels were decreased by 11% and HDL-C levels were increased by 10% in the hormone treated group. Despite these changes, there were no significant differences between the groups in the primary outcome (nonfatal myocardial infarction or CHD death) or in any of the secondary cardiovascular outcomes (coronary revascularization, unstable angina, congestive heart failure, resuscitated cardiac arrest, stroke or transient ischemic attack, and peripheral arterial disease). Interestingly, there were more CHD events in the hormone group in year 1 but fewer in years 4 and 5 compared to the placebo group. An unblinded extension of the HERS trial for an additional 2.7 years (HERSII) found that the lower rates of CHD events among women in the hormone group in the final years of HERS did not persist during additional years of follow-up. After 6.8 years, hormone therapy did not reduce the risk of cardiovascular events in women with pre-existing cardiovascular disease (428). As expected, hormone therapy decreased Lp(a) levels and in a post hoc analysis there was a suggestion that individuals with high baseline Lp(a) levels and individuals who had a robust decrease in Lp(a) with hormone therapy had a reduction in cardiovascular events (429). Of course, these results are not definitive and suggest the need for further focused trials of hormone therapy in postmenopausal women with elevated Lp(a) levels.

 

Women’s Health Initiative- Estrogen/Progesterone Therapy

 

The Women’s Health Initiative (WHI) examined the effect of hormone replacement therapy in women with and without an intact uterus. The WHI included a randomized primary-prevention trial of conjugated equine estrogens (CEE) (0.625 mg per day) plus continuous medroxyprogesterone acetate (MPA) (2.5 mg per day) or placebo in 16,608 postmenopausal women with an intact uterus who were 50 to 79 years of age at base line (430). As expected, hormone therapy lowered LDL-C levels by 12.7% and increased HDL-C levels by 7.3% and triglycerides by 6.9%. Despite these changes, after a mean follow-up of 5.2 years (planned duration, 8.5 years), the data and safety monitoring board recommended terminating the trial because the overall risks exceeded the benefits. Combined hormone therapy was associated with a hazard ratio for nonfatal myocardial infarction or death due to CHD of 1.24 in the hormone treated group. Similar to the HERS trial an increased risk of cardiovascular events was greatest in the first year of hormone therapy (HR 1.81).

 

Women’s Health Initiative- Estrogen Alone Therapy

 

In women without a uterus, the WHI carried out a randomized, double-blind, placebo-controlled trial of 0.625mg per day of conjugated equine estrogen (CEE) or placebo in 10,739 postmenopausal women, aged 50-79 years of age (431). As expected, the CEE group demonstrated a significant decrease in LDL-C compared to placebo group (−13.7% vs –1.0%, P<.001) and a much larger increase in HDL-C (15.1% vs 1.1%, P<.001). Additionally, large increases in triglyceride levels were observed in the CEE group (25.0% vs 3.0%, P<.001). After an average follow-up of 6.8 years, the estimated hazard ratio for nonfatal myocardial infarction or CHD death in the CEE vs placebo was 0.91 (0.75-1.12). However, the incidence of stroke was increased by 39% in the CEE group (P=.007).

 

These two initial reports of the results of the WHI coupled with the HERS trial indicated that hormone replacement therapy was not effective in reducing atherosclerotic cardiovascular disease events in a broad spectrum of postmenopausal women.

 

Women’s Health Initiative- Extension

 

In 2013 a report was published that extended the follow-up of both the estrogen alone and the combined estrogen/progesterone protocols of the WHI to 13 years (432). It should be noted that after the intervention phase ended only a very small percentage of subjects continued hormonal therapy (<4%). During the cumulative 13-year follow-up, the hazard ratios for nonfatal myocardial infarction or coronary death were 1.09 for CEE plus MPA and 0.94 for CEE alone compared with the placebo groups (both NS). During the 13-year follow-up the hazard ratios for stroke were higher in the hormone therapy groups compared with the placebo groups (HR, 1.16 for CEE plus MPA; HR, 1.15 for CEE alone). Although with cessation of hormonal therapy the risk of atherosclerotic cardiovascular disease appeared to diminish, due to the open label nature of this analysis these data are difficult to interpret.  Notably, there was no evidence for a “legacy effect” of cardiovascular benefit or harm after discontinuing hormone therapy. Thus, even with longer follow-up hormonal therapy did not demonstrate a reduction in atherosclerotic cardiovascular disease.

 

The Subject Age or Time Since Menopause Hypothesis

 

The WHI results, coupled with those of the HERS trial, have been translated into a recommendation that hormone replacement therapy not be used for cardiovascular disease prevention, that it not be started unless needed for postmenopausal symptom relief, and that it be terminated as soon as possible after obtaining symptom relief. This official interpretation is not accepted, however, by some gynecologists and lipidologists because studies have suggested a more nuanced approach (433). For example,  further analysis of the WHI results have suggested that age and/or time from menopause influences the effect of hormonal therapy on atherosclerotic cardiovascular disease events (432). In individuals 50-59 years of age who started hormone treatment with estrogen alone, there was a 40% reduction in coronary heart disease that was borderline statistically significant (p=0.08). In older individuals treated with estrogen alone there was no reduction or even a slight increase in coronary heart disease. In the 50-59 years of age group on estrogen alone, there was a 45% reduction in myocardial infarctions whereas in the 70-79 years of age group, there was a 24% increase in events. In the estrogen-progestin trial the age effect was not observed (Table 19).

 

Table 19. Effect of Age of Starting Hormone Therapy on Coronary Events in Women’s Health Initiative

Endpoint and Age at Study Entry

Estrogen-Progestin

Estrogen Alone

 

Relative Risk

Relative Risk

Coronary Heart Disease

 

 

50-59yrs

1.34

0.60

60-69yrs

1.01

0.95

70-79yrs

1.31

1.09

Myocardial Infarction

 

 

50-59yrs

1.32

0.55

60-69yrs

1.05

0.95

70-79yrs

1.46

1.24

Coronary Revascularization

 

 

50-59yrs

1.03

0.56

60-69yrs

0.85

1.13

70-79yrs

1.08

1.07

 

A separate, somewhat different age-subgroup analyses from the WHI showed an increase in both coronary heart disease and stroke only in women who started HRT after age 70, while in those age 60-70, there was an increase in stroke but no change in coronary heart disease (434).  In further contrast, in those who started hormone therapy before 60 there was no change in stroke, a trend towards decreased coronary heart disease in the CEE study, a trend towards improved global health index in the CEE study, and a statistically significant decrease in total mortality in both studies combined. In fact, there was a trend towards less harm and/or greater benefit in all major endpoints with decreasing age at treatment onset (434). 

 

The age effect is further supported by a meta-analysis of 23 trials with 39,049 women, which showed that hormone therapy significantly reduced CHD events in younger women (OR 0.68 [confidence interval (C I), 0.48 to 0.96]), but not in older women (OR 1.03 [CI, 0.91 to 1.16]) (435). A Cochrane meta-analysis also found an increased risk of cardiovascular events in older individuals treated with hormone therapy but those who started hormone therapy less than 10 years after the menopause had a decreased risk of coronary heart disease (RR 0.52, 95% CI 0.29 to 0.96) (436). Additionally, a more recent randomized trial in 1006 healthy women aged 45-58 who were recently postmenopausal demonstrated that hormonal therapy decreased an end point of death, myocardial infarction, or heart failure by 39% and myocardial infarction by 55% (437). The more clearly positive results may have been due to inclusion of younger women who were closer to the menopause (average 50 years of age and 0.7 years postmenopausal) than in the WHI study. Taken together, these results suggest that younger women who have recently undergone menopause may have either a decrease or no change in atherosclerotic cardiovascular disease when on hormonal therapy. In contrast, hormonal therapy in older women who have been postmenopausal for many years appears to increase the risk of atherosclerotic cardiovascular disease.

 

A possible explanation for the effect of age and/or time since menopause on the response to hormonal therapy could be the extent of underlying vascular disease (438). Younger women are more likely to have “healthy” vessels and in these circumstances hormonal therapy is beneficial. In contrast, in older women who may already have underlying atherosclerosis, treatment with hormonal therapy is not beneficial but rather may be harmful.  Further support for this hypothesis is provided by subgroup analyses in the WHI showing that women without risk factors for atherosclerosis appear to benefit from hormone therapy (439,440). For example, in women with LDL-C levels less than 130mg/dl or without the metabolic syndrome, hormone therapy is beneficial. However, in women with LDL-C levels greater than 130mg/dl or with the metabolic syndrome, hormone therapy increases the risk of coronary heart disease (Table 20). Furthermore, a high cardiovascular risk score identified women at a higher risk for cardiovascular events with hormone replacement therapy (441). 

 

Table 20. Effect of Baseline Risk Factors on Coronary Heart Disease Risk

 

Odds Ratio for Hormone Therapy Effect

P, interaction

LDL-C (mg/dl)

 

 

<130

0.66

0.03

>130

1.46

 

LDL/HDL ratio

 

 

<2.5

0.60

0.002

>2.5

1.73

 

Metabolic Syndrome

 

 

No

0.97

0.03

Yes

2.26

 

  

Apart from these considerations of age at treatment onset, there appears to be a strong temporal pattern of risk for cardiovascular disease relative to the time course of hormone therapy. In both the HERS and WHI studies an increase in cardiovascular events occurred during the first year of hormone therapy followed by a decrease with continued treatment (442). Interestingly, a similar temporal pattern was seen in the observational Nurses’ Health Study (443). One can speculate that the increase in coagulation factors induced by hormone therapy might account for this early increase in cardiovascular events. In a separate but related point, observational studies have shown worse outcomes for women who have stopped hormone therapy vs. those who have continued hormone therapy (421). There has never been a randomized trial of hormone therapy discontinuation vs. continuation of hormone therapy so in patients doing well on hormone therapy it is unclear whether stopping therapy will markedly affect the risk of cardiovascular disease.

 

EFFECT OF HORMONE THERAPY ON ATHEROSCLEROSIS

 

Given the absence of definitive results in the clinical outcome studies, further insights may be gained by examining studies of anatomical atherosclerotic changes. Several studies have explored the effect of hormonal therapy on the progression of atherosclerosis measured by quantitative coronary angiography, carotid intima-media thickness (CIMT), or coronary calcium scores (CAC). In patients with pre-existing coronary artery disease, hormone replacement therapy did not affect the progression of coronary atherosclerosis or CIMT (444-447).  Another study of healthy menopausal women aged 42 to 58 years between 6 and 36 months from last menses without prior CVD events who had a CAC score less than 50 Agatston units reported that CIMT and CAC changes were not significantly different in the hormonal or placebo groups (448). In contrast, in one study of women without pre-existing atherosclerotic disease, hormone replacement therapy slowed the rate of progression of CIMT (449). These observations support the clinical outcome studies that have shown that women with pre-existing atherosclerotic cardiovascular disease do not benefit from hormone therapy. In contrast, in women without pre-existing atherosclerotic cardiovascular disease, hormone therapy may be beneficial or neutral depending upon the particular study.

 

In the WHI estrogen alone trial, coronary artery calcium scores were measured in women between 50-59 years of age at study entry (448). The mean coronary-artery calcium score after trial completion was lower in women receiving estrogen therapy (83.1A) than in women receiving placebo (123.1A) (P = 0.02). This indicates that calcified-plaque burden in the coronary arteries was lower in younger women assigned to estrogen also supporting the hypothesis that estrogen therapy reduces the progression of atherosclerosis in women who are recently menopausal and do not have pre-existing atherosclerosis.

 

Hodis and colleagues randomly assigned 643 healthy postmenopausal women who were stratified according to time since menopause (<6 years [early post-menopause ] or ≥10 years [late post-menopause]) to receive either oral 17β-estradiol plus progesterone for 10 days of each 30-day cycle or placebo (450). In support of the WHI results, in women who were less than 6 years past menopause, the mean CIMT increased by 0.0078 mm per year in the placebo group versus 0.0044 mm per year in the estradiol group (P=0.008) while in women who were 10 or more years past menopause the rate of CIMT progression in the placebo and estradiol groups were similar. Coronary-artery calcium, total stenosis, and plaque did not differ significantly between the placebo group and either early or late postmenopausal group on hormonal therapy. Nevertheless, these observations suggest a difference in response to hormonal replacement therapy depending on duration of time since menopause.

 

In summary, in older women or women with pre-existing atherosclerosis, the data demonstrates that hormone therapy is not beneficial and is likely harmful. In younger women or women without pre-existing atherosclerosis studies suggest that hormone therapy is either modestly beneficial or neutral.

 

ORAL CONTRACEPTIVES

 

A Cochrane review has recently addressed the effect of oral contraceptives on atherosclerotic cardiovascular disease (451). They reported that oral contraceptive use did not increase the risk of myocardial infarction or ischemic stroke compared with non-users. The risks did not vary according to the generation of progestogen or according to progestogen type. However, the risk of myocardial infarction or ischemic stroke appeared to increase with higher doses of estrogen. The risk of myocardial infarction or ischemic stroke was only increased in women using oral contraceptives containing ≥ 50 µg of estrogen. In another meta-analysis of progesterone only contraceptives there did not appear to be an increase in the risk of myocardial infarctions (452). Additionally, another recent meta-analysis reported an increase in ischemic strokes but no increase in myocardial infarctions with oral contraceptive use (453). It should be noted that earlier meta-analyses have reported an increased risk of myocardial infarctions and ischemic strokes, which may be related to differences in the composition of the products and doses being used in the past in oral contraceptives (454,455). Thus, oral contraceptives with higher doses of estrogen likely increase cardiovascular disease risk.

 

POLYCYSTIC OVARY SYNDROME (PCOS)

 

A recent meta-analysis of five case-control studies and five cohort studies involving a total of 104,392 subjects found that PCOS was associated with a significant increased risk of cardiovascular disease (OR = 1.30) (456). Another smaller meta-analysis reported a 2-fold risk of arterial disease for patients with PCOS compared to women without PCOS (457). In contrast, in a study of cardiovascular events in 309 women with PCOS vs. 343 women without PCOS followed for a mean duration of 23.7 years an increase in cardiovascular disease was not observed (458). Of note the population of patients with PCOS in this study did not have diabetes or dyslipidemia and their BMI was only slightly greater than the controls (29.4 kg/m2 vs 28.3 kg/m2). In recent reviews it was noted that an increased prevalence of cardiovascular disease in women with PCOS has not been conclusively demonstrated (459,460). It has been proposed that the increased risk of cardiovascular disease in women with PCOS is mainly observed in women who are obese and/or have diabetes (461). A meta-analysis of studies comparing carotid intima-media thickness (CIMT) in individuals with PCOS vs. controls reported that women with PCOS have a higher mean CIMT compared with non-PCOS controls (462) but not all studies have shown this relationship (463).  Most but not all studies have shown that women with PCOS have higher coronary calcium scores than controls (463-468). In PCOS it is likely that many factors, such as decreased estrogen levels, increased testosterone levels, insulin resistance, hypertension, obesity, increased inflammation, alterations in glucose homeostasis, etc., could contribute to the increased cardiovascular risk in addition to a pro-atherogenic lipid profile and differences in the prevalence of various cardiovascular risk factors in patients with PCOS could account for the variable risk of cardiovascular events.

 

MANAGEMENT GUIDELINES

 

In 2020 the Endocrine Society published guidelines on the treatment of lipid disorders in patients with endocrine disorders (268). These recommendations are summarized in table 21.

 

Table 21. Endocrine Society Guidelines for the Management of Lipids in Patients with Endocrine Diseases

GH Deficiency

Obtain a lipid profile at diagnosis

GH deficiency associated with hypopituitarism

Assess and treat lipids and other cardiovascular risk factors

Acromegaly

Measure lipid profile before and after treatment of GH excess

Hypothyroidism

Suggest against treating hyperlipidemia until the patient is euthyroid

Subclinical hypothyroidism (TSH <10 mIU/L)

Suggest considering thyroxine treatment to reduce LDL-C levels

Hyperthyroid

Re-evaluate lipids after the patient becomes euthyroid

Cushing’s syndrome

Monitor the lipid profile

In adults with persistent endogenous Cushing syndrome, we suggest statin therapy (LDL-C > 70mg/dL), to reduce ASCVD risk, irrespective of the risk score

Hypogonadism

Testosterone as symptomatically indicated, and not to improve dyslipidemia or ASCVD risk.

Polycystic ovary syndrome

Obtaining a fasting lipid panel at diagnosis to assess ASCVD risk

Menopause and hormonal replacement

Treat dyslipidemia with statin therapy, rather than hormone therapy.

In women who enter menopause early (<40 to 45 years old), we recommend assessment and treatment of lipids and other ASCVD risk factors

Gender-affirming hormone therapy

In transwomen and transmen who have taken or are taking gender-affirming hormone therapy, assess ASCVD risk by guidelines for non-transgender adults

GH- Growth Hormone

 

ACKOWLEDGEMENTS

 

The author thanks Drs Carl Grunfeld and Eliot Brinton for their help in earlier editions of this chapter.

 

REFERENCES

 

  1. Pala NA, Laway BA, Misgar RA, Dar RA. Metabolic abnormalities in patients with prolactinoma: response to treatment with cabergoline. Diabetol Metab Syndr 2015; 7:99
  2. Pelkonen R, Nikkila EA, Grahne B. Serum lipids, postheparin plasma lipase activities and glucose tolerance in patients with prolactinoma. Clin Endocrinol (Oxf) 1982; 16:383-390
  3. Erem C, Kocak M, Nuhoglu I, Yilmaz M, Ucuncu O. Blood coagulation, fibrinolysis and lipid profile in patients with prolactinoma. Clin Endocrinol (Oxf) 2010; 73:502-507
  4. Oppenheim DS, Greenspan SL, Zervas NT, Schoenfeld DA, Klibanski A. Elevated serum lipids in hypogonadal men with and without hyperprolactinemia. Ann Intern Med 1989; 111:288-292
  5. Peric B, Kruljac I, Sundalic S, Pecina HI, Jovic A, Stefanovic M, Butorac D, Vrkljan M. Obesity and hypercholesterolemia in patients with prolactinomas: Could DHEA-S and growth hormone be the missing link? Endocr Res 2016; 41:200-206
  6. Berinder K, Nystrom T, Hoybye C, Hall K, Hulting AL. Insulin sensitivity and lipid profile in prolactinoma patients before and after normalization of prolactin by dopamine agonist therapy. Pituitary 2011; 14:199-207
  7. Fahy U, Hopton MI, Hartog M, Bolton CH, Hull MG. The lipoprotein profile of women with hyperprolactinaemic amenorrhoea. Hum Reprod 1999; 14:285-287
  8. Glezer A, Santana MR, Bronstein MD, Donato J, Jr., Jallad RS. The interplay between prolactin and cardiovascular disease. Front Endocrinol (Lausanne) 2022; 13:1018090
  9. Arslan MS, Topaloglu O, Sahin M, Tutal E, Gungunes A, Cakir E, Ozturk IU, Karbek B, Ucan B, Ginis Z, Cakal E, Ozbek M, Delibasi T. Preclinical atherosclerosis in patients with prolactinoma. Endocr Pract 2014; 20:447-451
  10. Jiang XB, Li CL, He DS, Mao ZG, Liu DH, Fan X, Hu B, Zhu YH, Wang HJ. Increased carotid intima media thickness is associated with prolactin levels in subjects with untreated prolactinoma: a pilot study. Pituitary2014; 17:232-239
  11. Heshmati HM, Turpin G, de Gennes JL. Chronic hyperprolactinemia and plasma lipids in women. Klin Wochenschr 1987; 65:516-519
  12. Ling C, Svensson L, Oden B, Weijdegard B, Eden B, Eden S, Billig H. Identification of functional prolactin (PRL) receptor gene expression: PRL inhibits lipoprotein lipase activity in human white adipose tissue. J Clin Endocrinol Metab 2003; 88:1804-1808
  13. Auriemma RS, Granieri L, Galdiero M, Simeoli C, Perone Y, Vitale P, Pivonello C, Negri M, Mannarino T, Giordano C, Gasperi M, Colao A, Pivonello R. Effect of cabergoline on metabolism in prolactinomas. Neuroendocrinology 2013; 98:299-310
  14. Ciresi A, Amato MC, Guarnotta V, Lo Castro F, Giordano C. Higher doses of cabergoline further improve metabolic parameters in patients with prolactinoma regardless of the degree of reduction in prolactin levels. Clin Endocrinol (Oxf) 2013; 79:845-852
  15. dos Santos Silva CM, Barbosa FR, Lima GA, Warszawski L, Fontes R, Domingues RC, Gadelha MR. BMI and metabolic profile in patients with prolactinoma before and after treatment with dopamine agonists. Obesity (Silver Spring) 2011; 19:800-805
  16. Inancli SS, Usluogullari A, Ustu Y, Caner S, Tam AA, Ersoy R, Cakir B. Effect of cabergoline on insulin sensitivity, inflammation, and carotid intima media thickness in patients with prolactinoma. Endocrine 2013; 44:193-199
  17. Byberg S, Futtrup J, Andreassen M, Krogh J. Metabolic effects of dopamine agonists in patients with prolactinomas: a systematic review and meta-analysis. Endocr Connect 2019;
  18. Schwetz V, Librizzi R, Trummer C, Theiler G, Stiegler C, Pieber TR, Obermayer-Pietsch B, Pilz S. Treatment of hyperprolactinaemia reduces total cholesterol and LDL in patients with prolactinomas. Metab Brain Dis2017; 32:155-161
  19. Khalil G, Khan FA, Jamal QM, Saleem A, Masroor H, Abbas K. Change in Insulin Sensitivity and Lipid Profile After Dopamine Agonist Therapy in Patients With Prolactinoma. Cureus 2021; 13:e17824
  20. Kamath V, Jones CN, Yip JC, Varasteh BB, Cincotta AH, Reaven GM, Chen YD. Effects of a quick-release form of bromocriptine (Ergoset) on fasting and postprandial plasma glucose, insulin, lipid, and lipoprotein concentrations in obese nondiabetic hyperinsulinemic women. Diabetes Care 1997; 20:1697-1701
  21. Pirchio R, Auriemma RS, Solari D, Arnesi M, Pivonello C, Negri M, de Angelis C, Cavallo LM, Cappabianca P, Colao A, Pivonello R. Effects of Pituitary Surgery and High-Dose Cabergoline Therapy on Metabolic Profile in Patients With Prolactinoma Resistant to Conventional Cabergoline Treatment. Front Endocrinol (Lausanne)2021; 12:769744
  22. Yazici D, Sunbul M, Yasar M, Deyneli O, Yavuz D. Is there an increased cardiovascular risk in patients with prolactinoma? A challenging question. J Clin Ultrasound 2021; 49:870-877
  23. Haring R, Friedrich N, Volzke H, Vasan RS, Felix SB, Dorr M, Meyer zu Schwabedissen HE, Nauck M, Wallaschofski H. Positive association of serum prolactin concentrations with all-cause and cardiovascular mortality. Eur Heart J 2014; 35:1215-1221
  24. Carrero JJ, Kyriazis J, Sonmez A, Tzanakis I, Qureshi AR, Stenvinkel P, Saglam M, Stylianou K, Yaman H, Taslipinar A, Vural A, Gok M, Yenicesu M, Daphnis E, Yilmaz MI. Prolactin levels, endothelial dysfunction, and the risk of cardiovascular events and mortality in patients with CKD. Clin J Am Soc Nephrol 2012; 7:207-215
  25. Krogh J, Selmer C, Torp-Pedersen C, Gislason GH, Kistorp C. Hyperprolactinemia and the Association with All-Cause Mortality and Cardiovascular Mortality. Horm Metab Res 2017; 49:411-417
  26. Toulis KA, Robbins T, Reddy N, Balachandran K, Gokhale K, Wijesinghe H, Cheng KK, Karavitaki N, Wass J, Nirantharakumar K. Males with prolactinoma are at increased risk of incident cardiovascular disease. Clin Endocrinol (Oxf) 2018; 88:71-76
  27. Rosen T, Eden S, Larson G, Wilhelmsen L, Bengtsson BA. Cardiovascular risk factors in adult patients with growth hormone deficiency. Acta Endocrinol (Copenh) 1993; 129:195-200
  28. de Boer H, Blok GJ, Voerman HJ, Phillips M, Schouten JA. Serum lipid levels in growth hormone-deficient men. Metabolism 1994; 43:199-203
  29. Attanasio AF, Lamberts SW, Matranga AM, Birkett MA, Bates PC, Valk NK, Hilsted J, Bengtsson BA, Strasburger CJ. Adult growth hormone (GH)-deficient patients demonstrate heterogeneity between childhood onset and adult onset before and during human GH treatment. Adult Growth Hormone Deficiency Study Group. J Clin Endocrinol Metab 1997; 82:82-88
  30. Abdu TA, Neary R, Elhadd TA, Akber M, Clayton RN. Coronary risk in growth hormone deficient hypopituitary adults: increased predicted risk is due largely to lipid profile abnormalities. Clin Endocrinol (Oxf) 2001; 55:209-216
  31. Rizzo M, Trepp R, Berneis K, Christ ER. Atherogenic lipoprotein phenotype and low-density lipoprotein size and subclasses in patients with growth hormone deficiency before and after short-term replacement therapy. Eur J Endocrinol 2007; 156:361-367
  32. Thomas JD, Monson JP. Adult GH deficiency throughout lifetime. Eur J Endocrinol 2009; 161 Suppl 1:S97-S106
  33. Murray RD, Wieringa G, Lawrance JA, Adams JE, Shalet SM. Partial growth hormone deficiency is associated with an adverse cardiovascular risk profile and increased carotid intima-medial thickness. Clin Endocrinol (Oxf)2010; 73:508-515
  34. Abs R, Feldt-Rasmussen U, Mattsson AF, Monson JP, Bengtsson BA, Goth MI, Wilton P, Koltowska-Haggstrom M. Determinants of cardiovascular risk in 2589 hypopituitary GH-deficient adults - a KIMS database analysis. Eur J Endocrinol 2006; 155:79-90
  35. Salman S, Uzum AK, Telci A, Alagol F, Ozbey NC. Serum adipokines and low density lipoprotein subfraction profile in hypopituitary patients with growth hormone deficiency. Pituitary 2012; 15:386-392
  36. O'Neal D, Hew FL, Sikaris K, Ward G, Alford F, Best JD. Low density lipoprotein particle size in hypopituitary adults receiving conventional hormone replacement therapy. J Clin Endocrinol Metab 1996; 81:2448-2454
  37. Feingold KR, Grunfeld C. Obesity and Dyslipidemia. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  38. Newman CB, Carmichael JD, Kleinberg DL. Effects of low dose versus high dose human growth hormone on body composition and lipids in adults with GH deficiency: a meta-analysis of placebo-controlled randomized trials. Pituitary 2015; 18:297-305
  39. Maison P, Griffin S, Nicoue-Beglah M, Haddad N, Balkau B, Chanson P, Metaanalysis of Blinded RP-CT. Impact of growth hormone (GH) treatment on cardiovascular risk factors in GH-deficient adults: a Metaanalysis of Blinded, Randomized, Placebo-Controlled Trials. J Clin Endocrinol Metab 2004; 89:2192-2199
  40. Gazzaruso C, Gola M, Karamouzis I, Giubbini R, Giustina A. Cardiovascular risk in adult patients with growth hormone (GH) deficiency and following substitution with GH--an update. J Clin Endocrinol Metab 2014; 99:18-29
  41. Eden S, Wiklund O, Oscarsson J, Rosen T, Bengtsson BA. Growth hormone treatment of growth hormone-deficient adults results in a marked increase in Lp(a) and HDL cholesterol concentrations. Arterioscler Thromb1993; 13:296-301
  42. Elbornsson M, Gotherstrom G, Bosaeus I, Bengtsson BA, Johannsson G, Svensson J. Fifteen years of GH replacement improves body composition and cardiovascular risk factors. Eur J Endocrinol 2013; 168:745-753
  43. Monson JP, Jonsson P, Koltowska-Haggstrom M, Kourides I. Growth hormone (GH) replacement decreases serum total and LDL-cholesterol in hypopituitary patients on maintenance HMG CoA reductase inhibitor (statin) therapy. Clin Endocrinol (Oxf) 2007; 67:623-628
  44. Bengtsson BA, Abs R, Bennmarker H, Monson JP, Feldt-Rasmussen U, Hernberg-Stahl E, Westberg B, Wilton P, Wuster C. The effects of treatment and the individual responsiveness to growth hormone (GH) replacement therapy in 665 GH-deficient adults. KIMS Study Group and the KIMS International Board. J Clin Endocrinol Metab 1999; 84:3929-3935
  45. Garry P, Collins P, Devlin JG. An open 36-month study of lipid changes with growth hormone in adults: lipid changes following replacement of growth hormone in adult acquired growth hormone deficiency. Eur J Endocrinol 1996; 134:61-66
  46. Laron Z, Wang XL, Klinger B, Silbergeld A, Wilcken DE. Growth hormone increases and insulin-like growth factor-I decreases circulating lipoprotein(a). Eur J Endocrinol 1997; 136:377-381
  47. O'Halloran DJ, Wieringa G, Tsatsoulis A, Shalet SM. Increased serum lipoprotein(a) concentrations after growth hormone (GH) treatment in patients with isolated GH deficiency. Ann Clin Biochem 1996; 33 ( Pt 4):330-334
  48. Weaver JU, Monson JP, Noonan K, John WG, Edwards A, Evans KA, Cunningham J. The effect of low dose recombinant human growth hormone replacement on regional fat distribution, insulin sensitivity, and cardiovascular risk factors in hypopituitary adults. J Clin Endocrinol Metab 1995; 80:153-159
  49. Fernandez-Perez L, Novoa J, Stahlberg N, Santana-Farre R, Boronat M, Marrero D, Henriquez-Hernandez L, Norstedt G, Flores-Morales A. The effect of in vivo growth hormone treatment on blood gene expression in adults with growth hormone deficiency reveals potential biomarkers to monitor growth hormone therapy. Clin Endocrinol (Oxf) 2010; 72:800-806
  50. Svensson J, Bengtsson BA, Taskinen MR, Wiklund O, Johannsson G. A nine-month, placebo-controlled study of the effects of growth hormone treatment on lipoproteins and LDL size in abdominally obese men. Growth Horm IGF Res 2000; 10:118-126
  51. Olivecrona H, Ericsson S, Berglund L, Angelin B. Increased concentrations of serum lipoprotein (a) in response to growth hormone treatment. BMJ 1993; 306:1726-1727
  52. Laron Z. Increase of serum lipoprotein (a), an adverse effect of growth hormone treatment. Growth Horm IGF Res 2022; 67:101503
  53. Olivecrona H, Johansson AG, Lindh E, Ljunghall S, Berglund L, Angelin B. Hormonal regulation of serum lipoprotein(a) levels. Contrasting effects of growth hormone and insulin-like growth factor-I. Arteriosclerosis, thrombosis, and vascular biology 1995; 15:847-849
  54. Parini P, Angelin B, Lobie PE, Norstedt G, Rudling M. Growth hormone specifically stimulates the expression of low density lipoprotein receptors in human hepatoma cells. Endocrinology 1995; 136:3767-3773
  55. Rudling M, Norstedt G, Olivecrona H, Reihner E, Gustafsson JA, Angelin B. Importance of growth hormone for the induction of hepatic low density lipoprotein receptors. Proc Natl Acad Sci U S A 1992; 89:6983-6987
  56. Persson L, Cao G, Stahle L, Sjoberg BG, Troutt JS, Konrad RJ, Galman C, Wallen H, Eriksson M, Hafstrom I, Lind S, Dahlin M, Amark P, Angelin B, Rudling M. Circulating proprotein convertase subtilisin kexin type 9 has a diurnal rhythm synchronous with cholesterol synthesis and is reduced by fasting in humans. Arterioscler Thromb Vasc Biol 2010; 30:2666-2672
  57. Lind S, Rudling M, Ericsson S, Olivecrona H, Eriksson M, Borgstrom B, Eggertsen G, Berglund L, Angelin B. Growth hormone induces low-density lipoprotein clearance but not bile acid synthesis in humans. Arterioscler Thromb Vasc Biol 2004; 24:349-356
  58. Christ ER, Cummings MH, Jackson N, Stolinski M, Lumb PJ, Wierzbicki AS, Sonksen PH, Russell-Jones DL, Umpleby AM. Effects of growth hormone (GH) replacement therapy on low-density lipoprotein apolipoprotein B100 kinetics in adult patients with GH deficiency: a stable isotope study. J Clin Endocrinol Metab 2004; 89:1801-1807
  59. Cummings MH, Christ E, Umpleby AM, Albany E, Wierzbicki A, Lumb PJ, Sonksen PH, Russell-Jones DL. Abnormalities of very low density lipoprotein apolipoprotein B-100 metabolism contribute to the dyslipidaemia of adult growth hormone deficiency. J Clin Endocrinol Metab 1997; 82:2010-2013
  60. Christ ER, Cummings MH, Albany E, Umpleby AM, Lumb PJ, Wierzbicki AS, Naoumova RP, Boroujerdi MA, Sonksen PH, Russell-Jones DL. Effects of growth hormone (GH) replacement therapy on very low density lipoprotein apolipoprotein B100 kinetics in patients with adult GH deficiency: a stable isotope study. J Clin Endocrinol Metab 1999; 84:307-316
  61. Moller N, Vendelbo MH, Kampmann U, Christensen B, Madsen M, Norrelund H, Jorgensen JO. Growth hormone and protein metabolism. Clin Nutr 2009; 28:597-603
  62. Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocrine reviews 2009; 30:152-177
  63. Tao R, Acquati F, Marcovina SM, Hobbs HH. Human growth hormone increases apo(a) expression in transgenic mice. Arterioscler Thromb Vasc Biol 1999; 19:2439-2447
  64. Svensson J, Bengtsson BA, Rosen T, Oden A, Johannsson G. Malignant disease and cardiovascular morbidity in hypopituitary adults with or without growth hormone replacement therapy. J Clin Endocrinol Metab 2004; 89:3306-3312
  65. Bates AS, Van't Hoff W, Jones PJ, Clayton RN. The effect of hypopituitarism on life expectancy. J Clin Endocrinol Metab 1996; 81:1169-1172
  66. Erfurth EM, Bulow B, Hagmar LE. Is vascular mortality increased in hypopituitarism? Pituitary 2000; 3:77-81
  67. Bulow B, Hagmar L, Mikoczy Z, Nordstrom CH, Erfurth EM. Increased cerebrovascular mortality in patients with hypopituitarism. Clin Endocrinol (Oxf) 1997; 46:75-81
  68. Tomlinson JW, Holden N, Hills RK, Wheatley K, Clayton RN, Bates AS, Sheppard MC, Stewart PM. Association between premature mortality and hypopituitarism. West Midlands Prospective Hypopituitary Study Group. Lancet 2001; 357:425-431
  69. Stochholm K, Laursen T, Green A, Laurberg P, Andersen M, Kristensen LO, Feldt-Rasmussen U, Christiansen JS, Frydenberg M, Gravholt CH. Morbidity and GH deficiency: a nationwide study. Eur J Endocrinol 2008; 158:447-457
  70. Cannavo S, Marini F, Curto L, Torre ML, de Gregorio C, Salamone I, Alibrandi A, Trimarchi F. High prevalence of coronary calcifications and increased risk for coronary heart disease in adults with growth hormone deficiency. J Endocrinol Invest 2011; 34:32-37
  71. Colao A, Di Somma C, Spiezia S, Savastano S, Rota F, Savanelli MC, Lombardi G. Growth hormone treatment on atherosclerosis: results of a 5-year open, prospective, controlled study in male patients with severe growth hormone deficiency. J Clin Endocrinol Metab 2008; 93:3416-3424
  72. Markussis V, Beshyah SA, Fisher C, Sharp P, Nicolaides AN, Johnston DG. Detection of premature atherosclerosis by high-resolution ultrasonography in symptom-free hypopituitary adults. Lancet 1992; 340:1188-1192
  73. Benedini S, Dalle Carbonare L, Albiger N, Scanarini M, Bilora F, Petrobelli F, Giannini S, Mantero F, Scaroni C. Effect of short-term therapy with recombinant human growth hormone (GH) on metabolic parameters and preclinical atherosclerotic markers in hypopituitary patients with growth hormone deficiency. Horm Metab Res2006; 38:16-21
  74. Colao A, Di Somma C, Cuocolo A, Spinelli L, Acampa W, Spiezia S, Rota F, Savanelli MC, Lombardi G. Does a gender-related effect of growth hormone (GH) replacement exist on cardiovascular risk factors, cardiac morphology, and performance and atherosclerosis? Results of a two-year open, prospective study in young adult men and women with severe GH deficiency. J Clin Endocrinol Metab 2005; 90:5146-5155
  75. Soares DV, Spina LD, de Lima Oliveira Brasil RR, da Silva EM, Lobo PM, Salles E, Coeli CM, Conceicao FL, Vaisman M. Carotid artery intima-media thickness and lipid profile in adults with growth hormone deficiency after long-term growth hormone replacement. Metabolism 2005; 54:321-329
  76. Murata M, Kaji H, Mizuno I, Sakurai T, Iida K, Okimura Y, Chihara K. A study of carotid intima-media thickness in GH-deficient Japanese adults during onset among adults and children. Eur J Endocrinol 2003; 148:333-338
  77. Pfeifer M, Verhovec R, Zizek B, Prezelj J, Poredos P, Clayton RN. Growth hormone (GH) treatment reverses early atherosclerotic changes in GH-deficient adults. J Clin Endocrinol Metab 1999; 84:453-457
  78. Bengtsson BA, Koppeschaar HP, Abs R, Bennmarker H, Hernberg-Stahl E, Westberg B, Wilton P, Monson JP, Feldt-Rasmussen U, Wuster C. Growth hormone replacement therapy is not associated with any increase in mortality. KIMS Study Group. J Clin Endocrinol Metab 1999; 84:4291-4292
  79. Holmer H, Svensson J, Rylander L, Johannsson G, Rosen T, Bengtsson BA, Thoren M, Hoybye C, Degerblad M, Bramnert M, Hagg E, Eden Engstrom B, Ekman B, Norrving B, Hagmar L, Erfurth EM. Nonfatal stroke, cardiac disease, and diabetes mellitus in hypopituitary patients on hormone replacement including growth hormone. J Clin Endocrinol Metab 2007; 92:3560-3567
  80. van Bunderen CC, van Nieuwpoort IC, Arwert LI, Heymans MW, Franken AA, Koppeschaar HP, van der Lely AJ, Drent ML. Does growth hormone replacement therapy reduce mortality in adults with growth hormone deficiency? Data from the Dutch National Registry of Growth Hormone Treatment in adults. J Clin Endocrinol Metab 2011; 96:3151-3159
  81. Borson-Chazot F, Serusclat A, Kalfallah Y, Ducottet X, Sassolas G, Bernard S, Labrousse F, Pastene J, Sassolas A, Roux Y, Berthezene F. Decrease in carotid intima-media thickness after one year growth hormone (GH) treatment in adults with GH deficiency. J Clin Endocrinol Metab 1999; 84:1329-1333
  82. Cenci MC, Conceicao FL, Soares DV, Spina LD, Brasil RR, Lobo PM, Michmacher E, Vaisman M. Impact of 5 years of growth hormone replacement therapy on cardiovascular risk factors in growth hormone-deficient adults. Metabolism 2008; 57:121-129
  83. Gibney J, Wallace JD, Spinks T, Schnorr L, Ranicar A, Cuneo RC, Lockhart S, Burnand KG, Salomon F, Sonksen PH, Russell-Jones D. The effects of 10 years of recombinant human growth hormone (GH) in adult GH-deficient patients. J Clin Endocrinol Metab 1999; 84:2596-2602
  84. Oliveira JL, Aguiar-Oliveira MH, D'Oliveira A, Jr., Pereira RM, Oliveira CR, Farias CT, Barreto-Filho JA, Anjos-Andrade FD, Marques-Santos C, Nascimento-Junior AC, Alves EO, Oliveira FT, Campos VC, Ximenes R, Blackford A, Parmigiani G, Salvatori R. Congenital growth hormone (GH) deficiency and atherosclerosis: effects of GH replacement in GH-naive adults. J Clin Endocrinol Metab 2007; 92:4664-4670
  85. Vilar L, Naves LA, Costa SS, Abdalla LF, Coelho CE, Casulari LA. Increase of classic and nonclassic cardiovascular risk factors in patients with acromegaly. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 2007; 13:363-372
  86. Takeda R, Tatami R, Ueda K, Sagara H, Nakabayashi H, Mabuchi H. The incidence and pathogenesis of hyperlipidaemia in 16 consecutive acromegalic patients. Acta endocrinologica 1982; 100:358-362
  87. Nikkila EA, Pelkonen R. Serum lipids in acromegaly. Metabolism: clinical and experimental 1975; 24:829-838
  88. Landin-Wilhelmsen K, Tengborn L, Wilhelmsen L, Bengtsson BA. Elevated fibrinogen levels decrease following treatment of acromegaly. Clinical endocrinology 1997; 46:69-74
  89. Colao A, Marzullo P, Lombardi G. Effect of a six-month treatment with lanreotide on cardiovascular risk factors and arterial intima-media thickness in patients with acromegaly. European journal of endocrinology / European Federation of Endocrine Societies 2002; 146:303-309
  90. Colao A, Spinelli L, Cuocolo A, Spiezia S, Pivonello R, di Somma C, Bonaduce D, Salvatore M, Lombardi G. Cardiovascular consequences of early-onset growth hormone excess. The Journal of clinical endocrinology and metabolism 2002; 87:3097-3104
  91. Colao A, Pivonello R, Grasso LF, Auriemma RS, Galdiero M, Savastano S, Lombardi G. Determinants of cardiac disease in newly diagnosed patients with acromegaly: results of a 10 year survey study. European journal of endocrinology / European Federation of Endocrine Societies 2011; 165:713-721
  92. Boero L, Manavela M, Gomez Rosso L, Insua C, Berardi V, Fornari MC, Brites F. Alterations in biomarkers of cardiovascular disease (CVD) in active acromegaly. Clinical endocrinology 2009; 70:88-95
  93. Boero L, Manavela M, Merono T, Maidana P, Gomez Rosso L, Brites F. GH levels and insulin sensitivity are differently associated with biomarkers of cardiovascular disease in active acromegaly. Clinical endocrinology2012; 77:579-585
  94. Arosio M, Sartore G, Rossi CM, Casati G, Faglia G, Manzato E. LDL physical properties, lipoprotein and Lp(a) levels in acromegalic patients. Effects of octreotide therapy. Italian Multicenter Octreotide Study Group. Atherosclerosis 2000; 151:551-557
  95. Beentjes JA, van Tol A, Sluiter WJ, Dullaart RP. Low plasma lecithin:cholesterol acyltransferase and lipid transfer protein activities in growth hormone deficient and acromegalic men: role in altered high density lipoproteins. Atherosclerosis 2000; 153:491-498
  96. Ciresi A, Amato MC, Pivonello R, Nazzari E, Grasso LF, Minuto F, Ferone D, Colao A, Giordano C. The metabolic profile in active acromegaly is gender-specific. The Journal of clinical endocrinology and metabolism2013; 98:E51-59
  97. Sesmilo G, Fairfield WP, Katznelson L, Pulaski K, Freda PU, Bonert V, Dimaraki E, Stavrou S, Vance ML, Hayden D, Klibanski A. Cardiovascular risk factors in acromegaly before and after normalization of serum IGF-I levels with the GH antagonist pegvisomant. J Clin Endocrinol Metab 2002; 87:1692-1699
  98. Parkinson C, Drake WM, Wieringa G, Yates AP, Besser GM, Trainer PJ. Serum lipoprotein changes following IGF-I normalization using a growth hormone receptor antagonist in acromegaly. Clin Endocrinol (Oxf) 2002; 56:303-311
  99. Maldonado Castro GF, Escobar-Morreale HF, Ortega H, Gomez-Coronado D, Balsa Barro JA, Varela C, Lasuncion MA. Effects of normalization of GH hypersecretion on lipoprotein(a) and other lipoprotein serum levels in acromegaly. Clinical endocrinology 2000; 53:313-319
  100. Berg C, Petersenn S, Lahner H, Herrmann BL, Buchfelder M, Droste M, Stalla GK, Strasburger CJ, Roggenbuck U, Lehmann N, Moebus S, Jockel KH, Mohlenkamp S, Erbel R, Saller B, Mann K, Investigative Group of the Heinz Nixdorf Recall S, the German Pegvisomant Observational Study B, Investigators. Cardiovascular risk factors in patients with uncontrolled and long-term acromegaly: comparison with matched data from the general population and the effect of disease control. The Journal of clinical endocrinology and metabolism 2010; 95:3648-3656
  101. Tan KC, Shiu SW, Janus ED, Lam KS. LDL subfractions in acromegaly: relation to growth hormone and insulin-like growth factor-I. Atherosclerosis 1997; 129:59-65
  102. Wildbrett J, Hanefeld M, Fucker K, Pinzer T, Bergmann S, Siegert G, Breidert M. Anomalies of lipoprotein pattern and fibrinolysis in acromegalic patients: relation to growth hormone levels and insulin-like growth factor I. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association 1997; 105:331-335
  103. Maffei P, Sicolo N, Plebani M. Lipoprotein(a) in acromegaly. Annals of internal medicine 1999; 130:537-538
  104. Lam KS, Pang RW, Janus ED, Kung AW, Wang CC. Serum apolipoprotein(a) correlates with growth hormone levels in Chinese patients with acromegaly. Atherosclerosis 1993; 104:183-188
  105. Tsuchiya H, Onishi T, Mogami H, Iida M. Lipid metabolism in acromegalic patients before and after selective pituitary adenomectomy. Endocrinologia japonica 1990; 37:797-807
  106. Tan KC, Pang RW, Tiu SC, Lam KS. Effects of treatment with Sandostatin LAR on small dense LDL and remnant-like lipoproteins in patients with acromegaly. Clinical endocrinology 2003; 59:558-564
  107. Reyes-Vidal C, Fernandez JC, Bruce JN, Crisman C, Conwell IM, Kostadinov J, Geer EB, Post KD, Freda PU. Prospective study of surgical treatment of acromegaly: effects on ghrelin, weight, adiposity, and markers of CV risk. The Journal of clinical endocrinology and metabolism 2014; 99:4124-4132
  108. Oscarsson J, Wiklund O, Jakobsson KE, Petruson B, Bengtsson BA. Serum lipoproteins in acromegaly before and 6-15 months after transsphenoidal adenomectomy. Clinical endocrinology 1994; 41:603-608
  109. James RA, Moller N, Chatterjee S, White M, Kendall-Taylor P. Carbohydrate tolerance and serum lipids in acromegaly before and during treatment with high dose octreotide. Diabetic medicine : a journal of the British Diabetic Association 1991; 8:517-523
  110. Cohen R, Chanson P, Bruckert E, Timsit J, Legrand A, Harris AG, Guillausseau PJ, Warnet A, Lubetzki J. Effects of octreotide on lipid metabolism in acromegaly. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 1992; 24:397-400
  111. Cozzolino A, Feola T, Simonelli I, Puliani G, Hasenmajer V, Minnetti M, Giannetta E, Gianfrilli D, Pasqualetti P, Lenzi A, Isidori AM. Metabolic complications in acromegaly after neurosurgery: a meta-analysis. Eur J Endocrinol 2020; 183:597-606
  112. Muller AF, Leebeek FW, Janssen JA, Lamberts SW, Hofland L, van der Lely AJ. Acute effect of pegvisomant on cardiovascular risk markers in healthy men: implications for the pathogenesis of atherosclerosis in GH deficiency. J Clin Endocrinol Metab 2001; 86:5165-5171
  113. Murase T, Yamada N, Ohsawa N, Kosaka K, Morita S, Yoshida S. Decline of postheparin plasma lipoprotein lipase in acromegalic patients. Metabolism: clinical and experimental 1980; 29:666-672
  114. Twickler TB, Dallinga-Thie GM, Zelissen PM, Koppeschaar HP, Erkelens DW. The atherogenic plasma remnant-like particle cholesterol concentration is increased in the fasting and postprandial state in active acromegalic patients. Clinical endocrinology 2001; 55:69-75
  115. Feingold KR, Grunfeld C. Introduction to Lipids and Lipoproteins. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  116. Mosca S, Paolillo S, Colao A, Bossone E, Cittadini A, Iudice FL, Parente A, Conte S, Rengo G, Leosco D, Trimarco B, Filardi PP. Cardiovascular involvement in patients affected by acromegaly: an appraisal. International journal of cardiology 2013; 167:1712-1718
  117. Colao A, Ferone D, Marzullo P, Lombardi G. Systemic complications of acromegaly: epidemiology, pathogenesis, and management. Endocrine reviews 2004; 25:102-152
  118. Schofl C, Petroff D, Tonjes A, Grussendorf M, Droste M, Stalla G, Jaursch-Hancke C, Stormann S, Schopohl J. Incidence of myocardial infarction and stroke in acromegaly patients: results from the German Acromegaly Registry. Pituitary 2017; 20:635-642
  119. Hong S, Kim KS, Han K, Park CY. Acromegaly and cardiovascular outcomes: a cohort study. Eur Heart J 2022; 43:1491-1499
  120. Brevetti G, Marzullo P, Silvestro A, Pivonello R, Oliva G, di Somma C, Lombardi G, Colao A. Early vascular alterations in acromegaly. The Journal of clinical endocrinology and metabolism 2002; 87:3174-3179
  121. Colao A, Spiezia S, Cerbone G, Pivonello R, Marzullo P, Ferone D, Di Somma C, Assanti AP, Lombardi G. Increased arterial intima-media thickness by B-M mode echodoppler ultrasonography in acromegaly. Clinical endocrinology 2001; 54:515-524
  122. De Martino MC, Auriemma RS, Brevetti G, Vitale G, Schiano V, Galdiero M, Grasso L, Lombardi G, Colao A, Pivonello R. The treatment with growth hormone receptor antagonist in acromegaly: effect on vascular structure and function in patients resistant to somatostatin analogues. Journal of endocrinological investigation2010; 33:663-670
  123. Kartal I, Oflaz H, Pamukcu B, Meric M, Aral F, Ozbey N, Alagol F. Investigation of early atherosclerotic changes in acromegalic patients. International journal of clinical practice 2010; 64:39-44
  124. Ozkan C, Altinova AE, Cerit ET, Yayla C, Sahinarslan A, Sahin D, Dincel AS, Toruner FB, Akturk M, Arslan M. Markers of early atherosclerosis, oxidative stress and inflammation in patients with acromegaly. Pituitary 2015; 18:621-629
  125. Otsuki M, Kasayama S, Yamamoto H, Saito H, Sumitani S, Kouhara H, Saitoh Y, Ohnishi T, Arita N. Characterization of premature atherosclerosis of carotid arteries in acromegalic patients. Clinical endocrinology2001; 54:791-796
  126. Boysan SN, Kantarci F, Celik O, Mihmanli I, Gazioglu N, Kadioglu P. Atherosclerotic risk factors and premature atherosclerosis in acromegaly before and after 48 months of octreotide-LAR treatment. Angiology 2012; 63:522-527
  127. Akutsu H, Kreutzer J, Wasmeier G, Ropers D, Rost C, Mohlig M, Wallaschofski H, Buchfelder M, Schofl C. Acromegaly per se does not increase the risk for coronary artery disease. European journal of endocrinology / European Federation of Endocrine Societies 2010; 162:879-886
  128. Bogazzi F, Battolla L, Spinelli C, Rossi G, Gavioli S, Di Bello V, Cosci C, Sardella C, Volterrani D, Talini E, Pepe P, Falaschi F, Mariani G, Martino E. Risk factors for development of coronary heart disease in patients with acromegaly: a five-year prospective study. The Journal of clinical endocrinology and metabolism 2007; 92:4271-4277
  129. Cannavo S, Almoto B, Cavalli G, Squadrito S, Romanello G, Vigo MT, Fiumara F, Benvenga S, Trimarchi F. Acromegaly and coronary disease: an integrated evaluation of conventional coronary risk factors and coronary calcifications detected by computed tomography. The Journal of clinical endocrinology and metabolism 2006; 91:3766-3772
  130. Dos Santos Silva CM, Lima GA, Volschan IC, Gottlieb I, Kasuki L, Neto LV, Gadelha MR. Low risk of coronary artery disease in patients with acromegaly. Endocrine 2015; 50:749-755
  131. Herrmann BL, Severing M, Schmermund A, Berg C, Budde T, Erbel R, Mann K. Impact of disease duration on coronary calcification in patients with acromegaly. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association 2009; 117:417-422
  132. Duntas LH, Brenta G. The effect of thyroid disorders on lipid levels and metabolism. Med Clin North Am 2012; 96:269-281
  133. Retnakaran R, Connelly PW, Goguen J. Unmasking of type III hyperlipoproteinemia by hypothyroidism: a dramatic illustration of altered lipoprotein metabolism in a postpartum woman. Endocr Pract 2005; 11:394-398
  134. Feussner G, Ziegler R. Expression of type III hyperlipoproteinaemia in a subject with secondary hypothyroidism bearing the apolipoprotein E2/2 phenotype. J Intern Med 1991; 230:183-186
  135. Lindner MA, Illingworth DR. Expression of type III hyperlipoproteinemia in an adolescent patient with hypothyroidism. J Pediatr 1988; 113:86-89
  136. Verdugo C, Perrot L, Ponsin G, Valentin C, Berthezene F. Time-course of alterations of high density lipoproteins (HDL) during thyroxine administration to hypothyroid women. Eur J Clin Invest 1987; 17:313-316
  137. de Bruin TW, van Barlingen H, van Linde-Sibenius Trip M, van Vuurst de Vries AR, Akveld MJ, Erkelens DW. Lipoprotein(a) and apolipoprotein B plasma concentrations in hypothyroid, euthyroid, and hyperthyroid subjects. J Clin Endocrinol Metab 1993; 76:121-126
  138. Klausen IC, Nielsen FE, Hegedus L, Gerdes LU, Charles P, Faergeman O. Treatment of hypothyroidism reduces low-density lipoproteins but not lipoprotein(a). Metabolism 1992; 41:911-914
  139. Tzotzas T, Krassas GE, Konstantinidis T, Bougoulia M. Changes in lipoprotein(a) levels in overt and subclinical hypothyroidism before and during treatment. Thyroid 2000; 10:803-808
  140. Kaliaperumal R, William E, Selvam T, Krishnan SM. Relationship between Lipoprotein(a) and Thyroid Hormones in Hypothyroid Patients. J Clin Diagn Res 2014; 8:37-39
  141. Murase T, Arimoto S, Okubo M, Morinaga S. Significant reduction of elevated serum lipoprotein(a) concentrations during levo-thyroxine-replacement therapy in a hypothyroid patient. J Clin Lipidol 2012; 6:388-391
  142. O'Brien T, Dinneen SF, O'Brien PC, Palumbo PJ. Hyperlipidemia in patients with primary and secondary hypothyroidism. Mayo Clin Proc 1993; 68:860-866
  143. Valdemarsson S, Hedner P, Nilsson-Ehle P. Dyslipoproteinaemia in hypothyroidism of pituitary origin: effects of L-thyroxine substitution on lipoprotein lipase, hepatic lipase, and on plasma lipoproteins. Acta Endocrinol (Copenh) 1983; 103:192-197
  144. Ito M, Arishima T, Kudo T, Nishihara E, Ohye H, Kubota S, Fukata S, Amino N, Kuma K, Sasaki I, Hiraiwa T, Hanafusa T, Takamatsu J, Miyauchi A. Effect of levo-thyroxine replacement on non-high-density lipoprotein cholesterol in hypothyroid patients. J Clin Endocrinol Metab 2007; 92:608-611
  145. Jung KY, Ahn HY, Han SK, Park YJ, Cho BY, Moon MK. Association between thyroid function and lipid profiles, apolipoproteins, and high-density lipoprotein function. J Clin Lipidol 2017; 11:1347-1353
  146. Kotwal A, Cortes T, Genere N, Hamidi O, Jasim S, Newman CB, Prokop LJ, Murad MH, Alahdab F. Treatment of Thyroid Dysfunction and Serum Lipids: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab2020; 105
  147. Pearce EN. Update in lipid alterations in subclinical hypothyroidism. J Clin Endocrinol Metab 2012; 97:326-333
  148. Wiersinga WM. Adult Hypothyroidism. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  149. Chubb SA, Davis WA, Davis TM. Interactions among thyroid function, insulin sensitivity, and serum lipid concentrations: the Fremantle diabetes study. J Clin Endocrinol Metab 2005; 90:5317-5320
  150. Rugge JB, Bougatsos C, Chou R. Screening and treatment of thyroid dysfunction: an evidence review for the U.S. Preventive Services Task Force. Ann Intern Med 2015; 162:35-45
  151. Meier C, Staub JJ, Roth CB, Guglielmetti M, Kunz M, Miserez AR, Drewe J, Huber P, Herzog R, Muller B. TSH-controlled L-thyroxine therapy reduces cholesterol levels and clinical symptoms in subclinical hypothyroidism: a double blind, placebo-controlled trial (Basel Thyroid Study). J Clin Endocrinol Metab 2001; 86:4860-4866
  152. Ochs N, Auer R, Bauer DC, Nanchen D, Gussekloo J, Cornuz J, Rodondi N. Meta-analysis: subclinical thyroid dysfunction and the risk for coronary heart disease and mortality. Ann Intern Med 2008; 148:832-845
  153. Razvi S, Shakoor A, Vanderpump M, Weaver JU, Pearce SH. The influence of age on the relationship between subclinical hypothyroidism and ischemic heart disease: a metaanalysis. J Clin Endocrinol Metab2008; 93:2998-3007
  154. Rodondi N, den Elzen WP, Bauer DC, Cappola AR, Razvi S, Walsh JP, Asvold BO, Iervasi G, Imaizumi M, Collet TH, Bremner A, Maisonneuve P, Sgarbi JA, Khaw KT, Vanderpump MP, Newman AB, Cornuz J, Franklyn JA, Westendorp RG, Vittinghoff E, Gussekloo J, Thyroid Studies C. Subclinical hypothyroidism and the risk of coronary heart disease and mortality. JAMA 2010; 304:1365-1374
  155. Sun J, Yao L, Fang Y, Yang R, Chen Y, Yang K, Tian L. Relationship between Subclinical Thyroid Dysfunction and the Risk of Cardiovascular Outcomes: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Int J Endocrinol 2017; 2017:8130796
  156. Ning Y, Cheng YJ, Liu LJ, Sara JD, Cao ZY, Zheng WP, Zhang TS, Han HJ, Yang ZY, Zhang Y, Wang FL, Pan RY, Huang JL, Wu LL, Zhang M, Wei YX. What is the association of hypothyroidism with risks of cardiovascular events and mortality? A meta-analysis of 55 cohort studies involving 1,898,314 participants. BMC Med 2017; 15:21
  157. Garber JR, Cobin RH, Gharib H, Hennessey JV, Klein I, Mechanick JI, Pessah-Pollack R, Singer PA, Woeber KA, American Association of Clinical E, American Thyroid Association Taskforce on Hypothyroidism in A. Clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Endocr Pract 2012; 18:988-1028
  158. Zhao T, Chen B, Zhou Y, Wang X, Zhang Y, Wang H, Shan Z. Effect of levothyroxine on the progression of carotid intima-media thickness in subclinical hypothyroidism patients: a meta-analysis. BMJ Open 2017; 7:e016053
  159. Aziz M, Kandimalla Y, Machavarapu A, Saxena A, Das S, Younus A, Nguyen M, Malik R, Anugula D, Latif MA, Humayun C, Khan IM, Adus A, Rasool A, Veledar E, Nasir K. Effect of Thyroxin Treatment on Carotid Intima-Media Thickness (CIMT) Reduction in Patients with Subclinical Hypothyroidism (SCH): a Meta-Analysis of Clinical Trials. J Atheroscler Thromb 2017; 24:643-659
  160. Gao N, Zhang W, Zhang YZ, Yang Q, Chen SH. Carotid intima-media thickness in patients with subclinical hypothyroidism: a meta-analysis. Atherosclerosis 2013; 227:18-25
  161. Blum MR, Gencer B, Adam L, Feller M, Collet TH, da Costa BR, Moutzouri E, Dopheide J, Depairon M, Sykiotis GP, Kearney P, Gussekloo J, Westendorp R, Stott DJ, Bauer DC, Rodondi N. Impact of Thyroid Hormone Therapy on Atherosclerosis in the Elderly With Subclinical Hypothyroidism: A Randomized Trial. J Clin Endocrinol Metab 2018; 103:2988-2997
  162. Park YJ, Lee YJ, Choi SI, Chun EJ, Jang HC, Chang HJ. Impact of subclinical hypothyroidism on the coronary artery disease in apparently healthy subjects. Eur J Endocrinol 2011; 165:115-121
  163. Silva N, Santos O, Morais F, Gottlieb I, Hadlich M, Rothstein T, Tauil M, Veras N, Vaisman M, Teixeira Pde F. Subclinical hypothyroidism represents an additional risk factor for coronary artery calcification, especially in subjects with intermediate and high cardiovascular risk scores. Eur J Endocrinol 2014; 171:327-334
  164. Verma R, Verma A, Gupta P, Agrawal NK. Coronary artery calcium scoring is a better predictor of cardiac risk in subclinical hypothyroidism patients with low-risk Framingham score. Indian J Endocrinol Metab 2016; 20:756-762
  165. Rosario PWS, Calsolari MR. Subclinical Hypothyroidism with TSH>7 mIU/l and</=10 mIU/l and Coronary Artery Disease. Horm Metab Res 2020; 52:85-88
  166. Walton KW, Scott PJ, Dykes PW, Davies JW. The significance of alterations in serum lipids in thyroid dysfunction. II. Alterations of the metabolism and turnover of 131-I-low-density lipoproteins in hypothyroidism and thyrotoxicosis. Clin Sci 1965; 29:217-238
  167. Chait A, Bierman EL, Albers JJ. Regulatory role of triiodothyronine in the degradation of low density lipoprotein by cultured human skin fibroblasts. J Clin Endocrinol Metab 1979; 48:887-889
  168. Staels B, Van Tol A, Chan L, Will H, Verhoeven G, Auwerx J. Alterations in thyroid status modulate apolipoprotein, hepatic triglyceride lipase, and low density lipoprotein receptor in rats. Endocrinology 1990; 127:1144-1152
  169. Salter AM, Hayashi R, al-Seeni M, Brown NF, Bruce J, Sorensen O, Atkinson EA, Middleton B, Bleackley RC, Brindley DN. Effects of hypothyroidism and high-fat feeding on mRNA concentrations for the low-density-lipoprotein receptor and on acyl-CoA:cholesterol acyltransferase activities in rat liver. Biochem J 1991; 276 ( Pt 3):825-832
  170. Scarabottolo L, Trezzi E, Roma P, Catapano AL. Experimental hypothyroidism modulates the expression of the low density lipoprotein receptor by the liver. Atherosclerosis 1986; 59:329-333
  171. Thompson GR, Soutar AK, Spengel FA, Jadhav A, Gavigan SJ, Myant NB. Defects of receptor-mediated low density lipoprotein catabolism in homozygous familial hypercholesterolemia and hypothyroidism in vivo. Proc Natl Acad Sci U S A 1981; 78:2591-2595
  172. Lopez D, Abisambra Socarras JF, Bedi M, Ness GC. Activation of the hepatic LDL receptor promoter by thyroid hormone. Biochim Biophys Acta 2007; 1771:1216-1225
  173. Shin DJ, Osborne TF. Thyroid hormone regulation and cholesterol metabolism are connected through Sterol Regulatory Element-Binding Protein-2 (SREBP-2). J Biol Chem 2003; 278:34114-34118
  174. Bonde Y, Breuer O, Lutjohann D, Sjoberg S, Angelin B, Rudling M. Thyroid hormone reduces PCSK9 and stimulates bile acid synthesis in humans. J Lipid Res 2014; 55:2408-2415
  175. Gong Y, Ma Y, Ye Z, Fu Z, Yang P, Gao B, Guo W, Hu D, Ye J, Ma S, Zhang F, Zhou L, Xu X, Li Z, Yang T, Zhou H. Thyroid stimulating hormone exhibits the impact on LDLR/LDL-c via up-regulating hepatic PCSK9 expression. Metabolism 2017; 76:32-41
  176. Goldberg IJ, Huang LS, Huggins LA, Yu S, Nagareddy PR, Scanlan TS, Ehrenkranz JR. Thyroid hormone reduces cholesterol via a non-LDL receptor-mediated pathway. Endocrinology 2012; 153:5143-5149
  177. Lin JZ, Martagon AJ, Hsueh WA, Baxter JD, Gustafsson JA, Webb P, Phillips KJ. Thyroid hormone receptor agonists reduce serum cholesterol independent of the LDL receptor. Endocrinology 2012; 153:6136-6144
  178. Day R, Gebhard RL, Schwartz HL, Strait KA, Duane WC, Stone BG, Oppenheimer JH. Time course of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase activity and messenger ribonucleic acid, biliary lipid secretion, and hepatic cholesterol content in methimazole-treated hypothyroid and hypophysectomized rats after triiodothyronine administration: possible linkage of cholesterol synthesis to biliary secretion. Endocrinology 1989; 125:459-468
  179. Pandak WM, Heuman DM, Redford K, Stravitz RT, Chiang JY, Hylemon PB, Vlahcevic ZR. Hormonal regulation of cholesterol 7alpha-hydroxylase specific activity, mRNA levels, and transcriptional activity in vivo in the rat. J Lipid Res 1997; 38:2483-2491
  180. Ness GC, Pendleton LC, Li YC, Chiang JY. Effect of thyroid hormone on hepatic cholesterol 7 alpha hydroxylase, LDL receptor, HMG-CoA reductase, farnesyl pyrophosphate synthetase and apolipoprotein A-I mRNA levels in hypophysectomized rats. Biochem Biophys Res Commun 1990; 172:1150-1156
  181. Galman C, Bonde Y, Matasconi M, Angelin B, Rudling M. Dramatically increased intestinal absorption of cholesterol following hypophysectomy is normalized by thyroid hormone. Gastroenterology 2008; 134:1127-1136
  182. Tancevski I, Demetz E, Eller P, Duwensee K, Hoefer J, Heim C, Stanzl U, Wehinger A, Auer K, Karer R, Huber J, Schgoer W, Van Eck M, Vanhoutte J, Fievet C, Stellaard F, Rudling M, Patsch JR, Ritsch A. The liver-selective thyromimetic T-0681 influences reverse cholesterol transport and atherosclerosis development in mice. PLoS One 2010; 5:e8722
  183. Kuusi T, Taskinen MR, Nikkila EA. Lipoproteins, lipolytic enzymes, and hormonal status in hypothyroid women at different levels of substitution. J Clin Endocrinol Metab 1988; 66:51-56
  184. Abrams JJ, Grundy SM, Ginsberg H. Metabolism of plasma triglycerides in hypothyroidism and hyperthyroidism in man. J Lipid Res 1981; 22:307-322
  185. Lam KS, Chan MK, Yeung RT. High-density lipoprotein cholesterol, hepatic lipase and lipoprotein lipase activities in thyroid dysfunction--effects of treatment. Q J Med 1986; 59:513-521
  186. Lithell H, Boberg J, Hellsing K, Ljunghall S, Lundqvist G, Vessby B, Wide L. Serum lipoprotein and apolipoprotein concentrations and tissue lipoprotein-lipase activity in overt and subclinical hypothyroidism: the effect of substitution therapy. Eur J Clin Invest 1981; 11:3-10
  187. Dimitriadis G, Mitrou P, Lambadiari V, Boutati E, Maratou E, Panagiotakos DB, Koukkou E, Tzanela M, Thalassinos N, Raptis SA. Insulin action in adipose tissue and muscle in hypothyroidism. J Clin Endocrinol Metab 2006; 91:4930-4937
  188. Valdemarsson S, Hedner P, Nilsson-Ehle P. Reversal of decreased hepatic lipase and lipoprotein lipase activities after treatment of hypothyroidism. Eur J Clin Invest 1982; 12:423-428
  189. Prieur X, Huby T, Coste H, Schaap FG, Chapman MJ, Rodriguez JC. Thyroid hormone regulates the hypotriglyceridemic gene APOA5. J Biol Chem 2005; 280:27533-27543
  190. Fugier C, Tousaint JJ, Prieur X, Plateroti M, Samarut J, Delerive P. The lipoprotein lipase inhibitor ANGPTL3 is negatively regulated by thyroid hormone. J Biol Chem 2006; 281:11553-11559
  191. Yang L, Yin R, Wang Z, Wang X, Zhang Y, Zhao D. Circulating Angptl3 and Angptl8 Are Increased in Patients with Hypothyroidism. Biomed Res Int 2019; 2019:3814687
  192. Fabbrini E, Magkos F, Patterson BW, Mittendorfer B, Klein S. Subclinical hypothyroidism and hyperthyroidism have opposite effects on hepatic very-low-density lipoprotein-triglyceride kinetics. J Clin Endocrinol Metab2012; 97:E414-418
  193. Tan KC, Shiu SW, Kung AW. Plasma cholesteryl ester transfer protein activity in hyper- and hypothyroidism. J Clin Endocrinol Metab 1998; 83:140-143
  194. Tan KC, Shiu SW, Kung AW. Effect of thyroid dysfunction on high-density lipoprotein subfraction metabolism: roles of hepatic lipase and cholesteryl ester transfer protein. J Clin Endocrinol Metab 1998; 83:2921-2924
  195. Ridgway ND, Dolphin PJ. Serum activity and hepatic secretion of lecithin:cholesterol acyltransferase in experimental hypothyroidism and hypercholesterolemia. J Lipid Res 1985; 26:1300-1313
  196. Ritter MC, Kannan CR, Bagdade JD. The effects of hypothyroidism and replacement therapy on cholesteryl ester transfer. J Clin Endocrinol Metab 1996; 81:797-800
  197. Dullaart RP, Hoogenberg K, Groener JE, Dikkeschei LD, Erkelens DW, Doorenbos H. The activity of cholesteryl ester transfer protein is decreased in hypothyroidism: a possible contribution to alterations in high-density lipoproteins. Eur J Clin Invest 1990; 20:581-587
  198. Packard CJ, Shepherd J, Lindsay GM, Gaw A, Taskinen MR. Thyroid replacement therapy and its influence on postheparin plasma lipases and apolipoprotein-B metabolism in hypothyroidism. J Clin Endocrinol Metab 1993; 76:1209-1216
  199. Pazos F, Alvarez JJ, Rubies-Prat J, Varela C, Lasuncion MA. Long-term thyroid replacement therapy and levels of lipoprotein(a) and other lipoproteins. J Clin Endocrinol Metab 1995; 80:562-566
  200. Johansson L, Rudling M, Scanlan TS, Lundasen T, Webb P, Baxter J, Angelin B, Parini P. Selective thyroid receptor modulation by GC-1 reduces serum lipids and stimulates steps of reverse cholesterol transport in euthyroid mice. Proc Natl Acad Sci U S A 2005; 102:10297-10302
  201. Boone LR, Lagor WR, Moya Mde L, Niesen MI, Rothblat GH, Ness GC. Thyroid hormone enhances the ability of serum to accept cellular cholesterol via the ABCA1 transporter. Atherosclerosis 2011; 218:77-82
  202. Heimberg M, Olubadewo JO, Wilcox HG. Plasma lipoproteins and regulation of hepatic metabolism of fatty acids in altered thyroid states. Endocr Rev 1985; 6:590-607
  203. Russo L, Nguyen TNH, Kyrilli A, Robin M, Bel Lassen P, Moreno-Reyes R, Corvilain B. Metabolic Changes after Radioiodine Correction of Grade 1 and Grade 2 Subclinical Hyperthyroidism. Eur Thyroid J 2021; 10:382-389
  204. The coronary drug project. Findings leading to further modifications of its protocol with respect to dextrothyroxine. The coronary drug project research group. JAMA 1972; 220:996-1008
  205. Angelin B, Rudling M. Lipid lowering with thyroid hormone and thyromimetics. Curr Opin Lipidol 2010; 21:499-506
  206. Bonde Y, Plosch T, Kuipers F, Angelin B, Rudling M. Stimulation of murine biliary cholesterol secretion by thyroid hormone is dependent on a functional ABCG5/G8 complex. Hepatology 2012; 56:1828-1837
  207. Stein EA. Low-density lipoprotein cholesterol reduction by inhibition of PCSK9. Curr Opin Lipidol 2013; 24:510-517
  208. Arnaldi G, Scandali VM, Trementino L, Cardinaletti M, Appolloni G, Boscaro M. Pathophysiology of dyslipidemia in Cushing's syndrome. Neuroendocrinology 2010; 92 Suppl 1:86-90
  209. Arnaldi G, Angeli A, Atkinson AB, Bertagna X, Cavagnini F, Chrousos GP, Fava GA, Findling JW, Gaillard RC, Grossman AB, Kola B, Lacroix A, Mancini T, Mantero F, Newell-Price J, Nieman LK, Sonino N, Vance ML, Giustina A, Boscaro M. Diagnosis and complications of Cushing's syndrome: a consensus statement. J Clin Endocrinol Metab 2003; 88:5593-5602
  210. Mancini T, Kola B, Mantero F, Boscaro M, Arnaldi G. High cardiovascular risk in patients with Cushing's syndrome according to 1999 WHO/ISH guidelines. Clin Endocrinol (Oxf) 2004; 61:768-777
  211. Giordano R, Picu A, Marinazzo E, D'Angelo V, Berardelli R, Karamouzis I, Forno D, Zinna D, Maccario M, Ghigo E, Arvat E. Metabolic and cardiovascular outcomes in patients with Cushing's syndrome of different aetiologies during active disease and 1 year after remission. Clin Endocrinol (Oxf) 2011; 75:354-360
  212. Krsek M, Silha JV, Jezkova J, Hana V, Marek J, Weiss V, Stepan JJ, Murphy LJ. Adipokine levels in Cushing's syndrome; elevated resistin levels in female patients with Cushing's syndrome. Clin Endocrinol (Oxf) 2004; 60:350-357
  213. Faggiano A, Pivonello R, Spiezia S, De Martino MC, Filippella M, Di Somma C, Lombardi G, Colao A. Cardiovascular risk factors and common carotid artery caliber and stiffness in patients with Cushing's disease during active disease and 1 year after disease remission. J Clin Endocrinol Metab 2003; 88:2527-2533
  214. Colao A, Pivonello R, Spiezia S, Faggiano A, Ferone D, Filippella M, Marzullo P, Cerbone G, Siciliani M, Lombardi G. Persistence of increased cardiovascular risk in patients with Cushing's disease after five years of successful cure. J Clin Endocrinol Metab 1999; 84:2664-2672
  215. Li Z, Zhang C, Geng C, Song Y. Metabolic profile differences in ACTH-dependent and ACTH-independent Cushing syndrome. Chronic Dis Transl Med 2022; 8:36-40
  216. Feingold KR, Grunfeld C. Diabetes and Dyslipidemia. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  217. Taskinen MR, Nikkila EA, Pelkonen R, Sane T. Plasma lipoproteins, lipolytic enzymes, and very low density lipoprotein triglyceride turnover in Cushing's syndrome. J Clin Endocrinol Metab 1983; 57:619-626
  218. Friedman TC, Mastorakos G, Newman TD, Mullen NM, Horton EG, Costello R, Papadopoulos NM, Chrousos GP. Carbohydrate and lipid metabolism in endogenous hypercortisolism: shared features with metabolic syndrome X and NIDDM. Endocr J 1996; 43:645-655
  219. Ettinger WH, Jr., Hazzard WR. Prednisone increases very low density lipoprotein and high density lipoprotein in healthy men. Metabolism 1988; 37:1055-1058
  220. Zimmerman J, Fainaru M, Eisenberg S. The effects of prednisone therapy on plasma lipoproteins and apolipoproteins: a prospective study. Metabolism 1984; 33:521-526
  221. Taskinen MR, Kuusi T, Yki-Jarvinen H, Nikkila EA. Short-term effects of prednisone on serum lipids and high density lipoprotein subfractions in normolipidemic healthy men. J Clin Endocrinol Metab 1988; 67:291-299
  222. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  223. de Guia RM, Herzig S. How Do Glucocorticoids Regulate Lipid Metabolism? Adv Exp Med Biol 2015; 872:127-144
  224. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid metabolism in humans: fuelling fat redistribution in the metabolic syndrome. J Endocrinol 2008; 197:189-204
  225. Hazra A, Pyszczynski NA, DuBois DC, Almon RR, Jusko WJ. Modeling of corticosteroid effects on hepatic low-density lipoprotein receptors and plasma lipid dynamics in rats. Pharm Res 2008; 25:769-780
  226. Galman C, Angelin B, Rudling M. Prolonged stimulation of the adrenals by corticotropin suppresses hepatic low-density lipoprotein and high-density lipoprotein receptors and increases plasma cholesterol. Endocrinology2002; 143:1809-1816
  227. Amatruda JM, Danahy SA, Chang CL. The effects of glucocorticoids on insulin-stimulated lipogenesis in primary cultures of rat hepatocytes. Biochem J 1983; 212:135-141
  228. Diamant S, Shafrir E. Modulation of the activity of insulin-dependent enzymes of lipogenesis by glucocorticoids. Eur J Biochem 1975; 53:541-546
  229. Krausz Y, Bar-On H, Shafrir E. Origin and pattern of glucocorticoid-induced hyperlipidemia in rats. Dose-dependent bimodal changes in serum lipids and lipoproteins in relation to hepatic lipogenesis and tissue lipoprotein lipase activity. Biochim Biophys Acta 1981; 663:69-82
  230. Mangiapane EH, Brindley DN. Effects of dexamethasone and insulin on the synthesis of triacylglycerols and phosphatidylcholine and the secretion of very-low-density lipoproteins and lysophosphatidylcholine by monolayer cultures of rat hepatocytes. Biochem J 1986; 233:151-160
  231. Lau DC, Roncari DA. Effects of glucocorticoid hormones on lipid-synthetic enzymes from different adipose tissue regions and from liver. Can J Biochem Cell Biol 1983; 61:1245-1250
  232. Pittner RA, Fears R, Brindley DN. Interactions of insulin, glucagon and dexamethasone in controlling the activity of glycerol phosphate acyltransferase and the activity and subcellular distribution of phosphatidate phosphohydrolase in cultured rat hepatocytes. Biochem J 1985; 230:525-534
  233. Dolinsky VW, Douglas DN, Lehner R, Vance DE. Regulation of the enzymes of hepatic microsomal triacylglycerol lipolysis and re-esterification by the glucocorticoid dexamethasone. Biochem J 2004; 378:967-974
  234. Wang CN, McLeod RS, Yao Z, Brindley DN. Effects of dexamethasone on the synthesis, degradation, and secretion of apolipoprotein B in cultured rat hepatocytes. Arterioscler Thromb Vasc Biol 1995; 15:1481-1491
  235. Cole TG, Wilcox HG, Heimberg M. Effects of adrenalectomy and dexamethasone on hepatic lipid metabolism. J Lipid Res 1982; 23:81-91
  236. Martin-Sanz P, Vance JE, Brindley DN. Stimulation of apolipoprotein secretion in very-low-density and high-density lipoproteins from cultured rat hepatocytes by dexamethasone. Biochem J 1990; 271:575-583
  237. Peckett AJ, Wright DC, Riddell MC. The effects of glucocorticoids on adipose tissue lipid metabolism. Metabolism 2011; 60:1500-1510
  238. Campbell JE, Peckett AJ, D'Souza A M, Hawke TJ, Riddell MC. Adipogenic and lipolytic effects of chronic glucocorticoid exposure. Am J Physiol Cell Physiol 2011; 300:C198-209
  239. Fain JN, Kovacev VP, Scow RO. Effect of growth hormone and dexamethasone on lipolysis and metabolism in isolated fat cells of the rat. J Biol Chem 1965; 240:3522-3529
  240. Samra JS, Clark ML, Humphreys SM, MacDonald IA, Bannister PA, Frayn KN. Effects of physiological hypercortisolemia on the regulation of lipolysis in subcutaneous adipose tissue. J Clin Endocrinol Metab 1998; 83:626-631
  241. Xu C, He J, Jiang H, Zu L, Zhai W, Pu S, Xu G. Direct effect of glucocorticoids on lipolysis in adipocytes. Mol Endocrinol 2009; 23:1161-1170
  242. Djurhuus CB, Gravholt CH, Nielsen S, Mengel A, Christiansen JS, Schmitz OE, Moller N. Effects of cortisol on lipolysis and regional interstitial glycerol levels in humans. Am J Physiol Endocrinol Metab 2002; 283:E172-177
  243. Divertie GD, Jensen MD, Miles JM. Stimulation of lipolysis in humans by physiological hypercortisolemia. Diabetes 1991; 40:1228-1232
  244. Slavin BG, Ong JM, Kern PA. Hormonal regulation of hormone-sensitive lipase activity and mRNA levels in isolated rat adipocytes. J Lipid Res 1994; 35:1535-1541
  245. Villena JA, Roy S, Sarkadi-Nagy E, Kim KH, Sul HS. Desnutrin, an adipocyte gene encoding a novel patatin domain-containing protein, is induced by fasting and glucocorticoids: ectopic expression of desnutrin increases triglyceride hydrolysis. J Biol Chem 2004; 279:47066-47075
  246. Birkenhager JC, Timmermans HA, Lamberts SW. Depressed plasma FFA turnover rate in Cushing's syndrome. J Clin Endocrinol Metab 1976; 42:28-32
  247. Saunders J, Hall SE, Sonksen PH. Glucose and free fatty acid turnover in Cushing's syndrome. J Endocrinol Invest 1980; 3:309-311
  248. Saladin R, Vu-Dac N, Fruchart JC, Auwerx J, Staels B. Transcriptional induction of rat liver apolipoprotein A-I gene expression by glucocorticoids requires the glucocorticoid receptor and a labile cell-specific protein. Eur J Biochem 1996; 239:451-459
  249. Taylor AH, Raymond J, Dionne JM, Romney J, Chan J, Lawless DE, Wanke IE, Wong NC. Glucocorticoid increases rat apolipoprotein A-I promoter activity. J Lipid Res 1996; 37:2232-2243
  250. Jansen H, van Tol A, Auwerx J, Skretting G, Staels B. Opposite regulation of hepatic lipase and lecithin: cholesterol acyltransferase by glucocorticoids in rats. Biochim Biophys Acta 1992; 1128:181-185
  251. Etxabe J, Vazquez JA. Morbidity and mortality in Cushing's disease: an epidemiological approach. Clin Endocrinol (Oxf) 1994; 40:479-484
  252. Graversen D, Vestergaard P, Stochholm K, Gravholt CH, Jorgensen JO. Mortality in Cushing's syndrome: a systematic review and meta-analysis. Eur J Intern Med 2012; 23:278-282
  253. Ntali G, Asimakopoulou A, Siamatras T, Komninos J, Vassiliadi D, Tzanela M, Tsagarakis S, Grossman AB, Wass JA, Karavitaki N. Mortality in Cushing's syndrome: systematic analysis of a large series with prolonged follow-up. Eur J Endocrinol 2013; 169:715-723
  254. Clayton RN, Jones PW, Reulen RC, Stewart PM, Hassan-Smith ZK, Ntali G, Karavitaki N, Dekkers OM, Pereira AM, Bolland M, Holdaway I, Lindholm J. Mortality in patients with Cushing's disease more than 10 years after remission: a multicentre, multinational, retrospective cohort study. Lancet Diabetes Endocrinol2016; 4:569-576
  255. van Haalen FM, Broersen LH, Jorgensen JO, Pereira AM, Dekkers OM. Management of endocrine disease: Mortality remains increased in Cushing's disease despite biochemical remission: a systematic review and meta-analysis. Eur J Endocrinol 2015; 172:R143-149
  256. Dekkers OM, Horvath-Puho E, Jorgensen JO, Cannegieter SC, Ehrenstein V, Vandenbroucke JP, Pereira AM, Sorensen HT. Multisystem morbidity and mortality in Cushing's syndrome: a cohort study. J Clin Endocrinol Metab 2013; 98:2277-2284
  257. Albiger N, Testa RM, Almoto B, Ferrari M, Bilora F, Petrobelli F, Pagnan A, Mantero F, Scaroni C. Patients with Cushing's syndrome have increased intimal media thickness at different vascular levels: comparison with a population matched for similar cardiovascular risk factors. Horm Metab Res 2006; 38:405-410
  258. Bassareo PP, Fanos V, Zaffanello M, Mercuro G. Early markers of cardiovascular dysfunction in young girls affected by Cushing's syndrome before and after successful cure. J Pediatr Endocrinol Metab 2010; 23:627-635
  259. Petramala L, Lorenzo D, Iannucci G, Concistre A, Zinnamosca L, Marinelli C, De Vincentis G, Ciardi A, De Toma G, Letizia C. Subclinical Atherosclerosis in Patients with Cushing Syndrome: Evaluation with Carotid Intima-Media Thickness and Ankle-Brachial Index. Endocrinol Metab (Seoul) 2015; 30:488-493
  260. Barahona MJ, Resmini E, Vilades D, Pons-Llado G, Leta R, Puig T, Webb SM. Coronary artery disease detected by multislice computed tomography in patients after long-term cure of Cushing's syndrome. J Clin Endocrinol Metab 2013; 98:1093-1099
  261. Neary NM, Booker OJ, Abel BS, Matta JR, Muldoon N, Sinaii N, Pettigrew RI, Nieman LK, Gharib AM. Hypercortisolism is associated with increased coronary arterial atherosclerosis: analysis of noninvasive coronary angiography using multidetector computerized tomography. J Clin Endocrinol Metab 2013; 98:2045-2052
  262. Fardet L, Petersen I, Nazareth I. Risk of cardiovascular events in people prescribed glucocorticoids with iatrogenic Cushing's syndrome: cohort study. BMJ 2012; 345:e4928
  263. Souverein PC, Berard A, Van Staa TP, Cooper C, Egberts AC, Leufkens HG, Walker BR. Use of oral glucocorticoids and risk of cardiovascular and cerebrovascular disease in a population based case-control study. Heart 2004; 90:859-865
  264. Wei L, MacDonald TM, Walker BR. Taking glucocorticoids by prescription is associated with subsequent cardiovascular disease. Ann Intern Med 2004; 141:764-770
  265. Varas-Lorenzo C, Rodriguez LA, Maguire A, Castellsague J, Perez-Gutthann S. Use of oral corticosteroids and the risk of acute myocardial infarction. Atherosclerosis 2007; 192:376-383
  266. Sharma ST, Nieman LK, Feelders RA. Comorbidities in Cushing's disease. Pituitary 2015; 18:188-194
  267. De Leo M, Pivonello R, Auriemma RS, Cozzolino A, Vitale P, Simeoli C, De Martino MC, Lombardi G, Colao A. Cardiovascular disease in Cushing's syndrome: heart versus vasculature. Neuroendocrinology 2010; 92 Suppl 1:50-54
  268. Newman CB, Blaha MJ, Boord JB, Cariou B, Chait A, Fein HG, Ginsberg HN, Goldberg IJ, Murad MH, Subramanian S, Tannock LR. Lipid Management in Patients with Endocrine Disorders: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2020; 105
  269. Gylling H, Vanhanen H, Miettinen TA. Effects of ketoconazole on cholesterol precursors and low density lipoprotein kinetics in hypercholesterolemia. J Lipid Res 1993; 34:59-67
  270. Miettinen TA. Cholesterol metabolism during ketoconazole treatment in man. J Lipid Res 1988; 29:43-51
  271. Gylling H, Vanhanen H, Miettinen TA. Hypolipidemic effect and mechanism of ketoconazole without and with cholestyramine in familial hypercholesterolemia. Metabolism 1991; 40:35-41
  272. Fleseriu M, Auchus RJ, Greenman Y, Zacharieva S, Geer EB, Salvatori R, Pivonello R, Feldt-Rasmussen U, Kennedy L, Buchfelder M, Biller BM, Cohen F, Heaney AP. Levoketoconazole treatment in endogenous Cushing's syndrome: extended evaluation of clinical, biochemical, and radiologic outcomes. Eur J Endocrinol2022; 187:859-871
  273. Feingold KR GC. Cholesterol Lowering Drugs. In: De Groot LJ B-PP, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, ed.

. South Dartmouth (MA: MDText.com, Inc; 2016.

  1. Maher VM, Trainer PJ, Scoppola A, Anderson JV, Thompson GR, Besser GM. Possible mechanism and treatment of o,p'DDD-induced hypercholesterolaemia. Q J Med 1992; 84:671-679
  2. Shawa H, Deniz F, Bazerbashi H, Hernandez M, Vassilopoulou-Sellin R, Jimenez C, Habra MA. Mitotane-induced hyperlipidemia: a retrospective cohort study. Int J Endocrinol 2013; 2013:624962
  3. Bianchini M, Puliani G, Chiefari A, Mormando M, Lauretta R, Appetecchia M. Metabolic and Endocrine Toxicities of Mitotane: A Systematic Review. Cancers (Basel) 2021; 13
  4. Stacpoole PW, Varnado CE, Island DP. Stimulation of rat liver 3-hydroxy-3-methylglutaryl-coenzyme A reductase activity by o,p'-DDD. Biochem Pharmacol 1982; 31:857-860
  5. Tada H, Nohara A, Kawashiri MA, Inazu A, Mabuchi H, Yamagishi M. Marked transient hypercholesterolemia caused by low-dose mitotane as adjuvant chemotherapy for adrenocortical carcinoma. J Atheroscler Thromb2014; 21:1326-1329
  6. Page ST, Krauss RM, Gross C, Ishida B, Heinecke JW, Tang C, Amory JK, Schaefer PM, Cox CJ, Kane J, Purnell JQ, Weinstein RL, Vaisar T. Impact of mifepristone, a glucocorticoid/progesterone antagonist, on HDL cholesterol, HDL particle concentration, and HDL function. J Clin Endocrinol Metab 2012; 97:1598-1605
  7. Ottosson M, Marin P, Karason K, Elander A, Bjorntorp P. Blockade of the glucocorticoid receptor with RU 486: effects in vitro and in vivo on human adipose tissue lipoprotein lipase activity. Obes Res 1995; 3:233-240
  8. Pivonello R, Arnaldi G, Scaroni C, Giordano C, Cannavo S, Iacuaniello D, Trementino L, Zilio M, Guarnotta V, Albani A, Cozzolino A, Michetti G, Boscaro M, Colao A. The medical treatment with pasireotide in Cushing's disease: an Italian multicentre experience based on "real-world evidence". Endocrine 2019; 64:657-672
  9. Albani A, Ferrau F, Ciresi A, Pivonello R, Scaroni C, Iacuaniello D, Zilio M, Guarnotta V, Alibrandi A, Messina E, Boscaro M, Giordano C, Colao A, Cannavo S. Pasireotide treatment reduces cardiometabolic risk in Cushing's disease patients: an Italian, multicenter study. Endocrine 2018; 61:118-124
  10. Pivonello R, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, Trovato A, Hughes G, Salgado LR, Lacroix A, Schopohl J, Biller BM, Pasireotide BSG. Pasireotide treatment significantly improves clinical signs and symptoms in patients with Cushing's disease: results from a Phase III study. Clin Endocrinol (Oxf)2014; 81:408-417
  11. Agledahl I, Skjaerpe PA, Hansen JB, Svartberg J. Low serum testosterone in men is inversely associated with non-fasting serum triglycerides: the Tromso study. Nutr Metab Cardiovasc Dis 2008; 18:256-262
  12. Haffner SM, Mykkanen L, Valdez RA, Katz MS. Relationship of sex hormones to lipids and lipoproteins in nondiabetic men. J Clin Endocrinol Metab 1993; 77:1610-1615
  13. Khaw KT, Barrett-Connor E. Endogenous sex hormones, high density lipoprotein cholesterol, and other lipoprotein fractions in men. Arterioscler Thromb 1991; 11:489-494
  14. Makinen JI, Perheentupa A, Irjala K, Pollanen P, Makinen J, Huhtaniemi I, Raitakari OT. Endogenous testosterone and serum lipids in middle-aged men. Atherosclerosis 2008; 197:688-693
  15. Van Pottelbergh I, Braeckman L, De Bacquer D, De Backer G, Kaufman JM. Differential contribution of testosterone and estradiol in the determination of cholesterol and lipoprotein profile in healthy middle-aged men. Atherosclerosis 2003; 166:95-102
  16. Hamalainen E, Adlercreutz H, Ehnholm C, Puska P. Relationships of serum lipoproteins and apoproteins to sex hormones and to the binding capacity of sex hormone binding globulin in healthy Finnish men. Metabolism1986; 35:535-541
  17. Wu FC, von Eckardstein A. Androgens and coronary artery disease. Endocr Rev 2003; 24:183-217
  18. Gyllenborg J, Rasmussen SL, Borch-Johnsen K, Heitmann BL, Skakkebaek NE, Juul A. Cardiovascular risk factors in men: The role of gonadal steroids and sex hormone-binding globulin. Metabolism 2001; 50:882-888
  19. Brand JS, van der Tweel I, Grobbee DE, Emmelot-Vonk MH, van der Schouw YT. Testosterone, sex hormone-binding globulin and the metabolic syndrome: a systematic review and meta-analysis of observational studies. Int J Epidemiol 2011; 40:189-207
  20. Corona G, Monami M, Rastrelli G, Aversa A, Tishova Y, Saad F, Lenzi A, Forti G, Mannucci E, Maggi M. Testosterone and metabolic syndrome: a meta-analysis study. J Sex Med 2011; 8:272-283
  21. Bann D, Wu FC, Keevil B, Lashen H, Adams J, Hardy R, Muniz G, Kuh D, Ben-Shlomo Y, Ong KK. Changes in testosterone related to body composition in late midlife: Findings from the 1946 British birth cohort study. Obesity (Silver Spring) 2015; 23:1486-1492
  22. Corona G, Monami M, Rastrelli G, Aversa A, Sforza A, Lenzi A, Forti G, Mannucci E, Maggi M. Type 2 diabetes mellitus and testosterone: a meta-analysis study. Int J Androl 2011; 34:528-540
  23. Corona G, Rastrelli G, Monami M, Saad F, Luconi M, Lucchese M, Facchiano E, Sforza A, Forti G, Mannucci E, Maggi M. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: a systematic review and meta-analysis. Eur J Endocrinol 2013; 168:829-843
  24. Sarwer DB, Spitzer JC, Wadden TA, Rosen RC, Mitchell JE, Lancaster K, Courcoulas A, Gourash W, Christian NJ. Sexual functioning and sex hormones in men who underwent bariatric surgery. Surg Obes Relat Dis 2015; 11:643-651
  25. Behre HM, Bockers A, Schlingheider A, Nieschlag E. Sustained suppression of serum LH, FSH and testosterone and increase of high-density lipoprotein cholesterol by daily injections of the GnRH antagonist cetrorelix over 8 days in normal men. Clin Endocrinol (Oxf) 1994; 40:241-248
  26. Mohamedali HZ, Breunis H, Timilshina N, Alibhai SM. Changes in blood glucose and cholesterol levels due to androgen deprivation therapy in men with non-metastatic prostate cancer. Can Urol Assoc J 2011; 5:28-32
  27. Saglam HS, Kose O, Kumsar S, Budak S, Adsan O. Fasting blood glucose and lipid profile alterations following twelve-month androgen deprivation therapy in men with prostate cancer. ScientificWorldJournal 2012; 2012:696329
  28. Smith MR, Finkelstein JS, McGovern FJ, Zietman AL, Fallon MA, Schoenfeld DA, Kantoff PW. Changes in body composition during androgen deprivation therapy for prostate cancer. J Clin Endocrinol Metab 2002; 87:599-603
  29. Eri LM, Urdal P, Bechensteen AG. Effects of the luteinizing hormone-releasing hormone agonist leuprolide on lipoproteins, fibrinogen and plasminogen activator inhibitor in patients with benign prostatic hyperplasia. J Urol1995; 154:100-104
  30. Bagatell CJ, Knopp RH, Vale WW, Rivier JE, Bremner WJ. Physiologic testosterone levels in normal men suppress high-density lipoprotein cholesterol levels. Ann Intern Med 1992; 116:967-973
  31. Yannucci J, Manola J, Garnick MB, Bhat G, Bubley GJ. The effect of androgen deprivation therapy on fasting serum lipid and glucose parameters. J Urol 2006; 176:520-525
  32. von Eckardstein A, Kliesch S, Nieschlag E, Chirazi A, Assmann G, Behre HM. Suppression of endogenous testosterone in young men increases serum levels of high density lipoprotein subclass lipoprotein A-I and lipoprotein(a). J Clin Endocrinol Metab 1997; 82:3367-3372
  33. Braga-Basaria M, Muller DC, Carducci MA, Dobs AS, Basaria S. Lipoprotein profile in men with prostate cancer undergoing androgen deprivation therapy. Int J Impot Res 2006; 18:494-498
  34. Torimoto K, Samma S, Kagebayashi Y, Chihara Y, Tanaka N, Hirayama A, Fujimoto K, Hirao Y. The effects of androgen deprivation therapy on lipid metabolism and body composition in Japanese patients with prostate cancer. Jpn J Clin Oncol 2011; 41:577-581
  35. Salvador C, Planas J, Agreda F, Placer J, Trilla E, Lopez MA, Morote J. Analysis of the lipid profile and atherogenic risk during androgen deprivation therapy in prostate cancer patients. Urol Int 2013; 90:41-44
  36. Cakici MC, Can B, Kazan O, Efiloglu O, Sendogan F, Ucar T, Turan T, Yildirim A. Effects of androgen deprivation therapy on metabolic markers and bioelectrical impedance analyze in prostate cancer patients. Rev Int Androl 2022; 20:96-101
  37. Whitsel EA, Boyko EJ, Matsumoto AM, Anawalt BD, Siscovick DS. Intramuscular testosterone esters and plasma lipids in hypogonadal men: a meta-analysis. Am J Med 2001; 111:261-269
  38. 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. Clin Endocrinol (Oxf) 2005; 63:280-293
  39. Fernandez-Balsells MM, Murad MH, Lane M, Lampropulos JF, Albuquerque F, Mullan RJ, Agrwal N, Elamin MB, Gallegos-Orozco JF, Wang AT, Erwin PJ, Bhasin S, Montori VM. Clinical review 1: Adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol Metab 2010; 95:2560-2575
  40. Haddad RM, Kennedy CC, Caples SM, Tracz MJ, Bolona ER, Sideras K, Uraga MV, Erwin PJ, Montori VM. Testosterone and cardiovascular risk in men: a systematic review and meta-analysis of randomized placebo-controlled trials. Mayo Clin Proc 2007; 82:29-39
  41. Corona G, Giagulli VA, Maseroli E, Vignozzi L, Aversa A, Zitzmann M, Saad F, Mannucci E, Maggi M. THERAPY OF ENDOCRINE DISEASE: Testosterone supplementation and body composition: results from a meta-analysis study. Eur J Endocrinol 2016; 174:R99-116
  42. Kumari K, Kumar R, Memon A, Kumari B, Tehrim M, Kumari P, Shehryar M, Islam H, Islam R, Khatri M, Kumar S, Kumar A. Treatment with Testosterone Therapy in Type 2 Diabetic Hypogonadal Adult Males: A Systematic Review and Meta-Analysis. Clin Pract 2023; 13:454-469
  43. Yassin A, Haider A, Haider KS, Caliber M, Doros G, Saad F, Garvey WT. Testosterone Therapy in Men With Hypogonadism Prevents Progression From Prediabetes to Type 2 Diabetes: Eight-Year Data From a Registry Study. Diabetes Care 2019; 42:1104-1111
  44. Schleich F, Legros JJ. Effects of androgen substitution on lipid profile in the adult and aging hypogonadal male. Eur J Endocrinol 2004; 151:415-424
  45. Saad F, Gooren LJ, Haider A, Yassin A. A dose-response study of testosterone on sexual dysfunction and features of the metabolic syndrome using testosterone gel and parenteral testosterone undecanoate. J Androl2008; 29:102-105
  46. Anderson RA, Wallace EM, Wu FC. Effect of testosterone enanthate on serum lipoproteins in man. Contraception 1995; 52:115-119
  47. Friedl KE, Hannan CJ, Jr., Jones RE, Plymate SR. High-density lipoprotein cholesterol is not decreased if an aromatizable androgen is administered. Metabolism 1990; 39:69-74
  48. Thompson PD, Cullinane EM, Sady SP, Chenevert C, Saritelli AL, Sady MA, Herbert PN. Contrasting effects of testosterone and stanozolol on serum lipoprotein levels. JAMA 1989; 261:1165-1168
  49. Mohler ER, 3rd, Ellenberg SS, Lewis CE, Wenger NK, Budoff MJ, Lewis MR, Barrett-Connor E, Swerdloff RS, Stephens-Shields A, Bhasin S, Cauley JA, Crandall JP, Cunningham GR, Ensrud KE, Gill TM, Matsumoto AM, Molitch ME, Pahor M, Preston PE, Hou X, Cifelli D, Snyder PJ. The Effect of Testosterone on Cardiovascular Biomarkers in the Testosterone Trials. J Clin Endocrinol Metab 2018; 103:681-688
  50. Webb OL, Laskarzewski PM, Glueck CJ. Severe depression of high-density lipoprotein cholesterol levels in weight lifters and body builders by self-administered exogenous testosterone and anabolic-androgenic steroids. Metabolism 1984; 33:971-975
  51. Hurley BF, Seals DR, Hagberg JM, Goldberg AC, Ostrove SM, Holloszy JO, Wiest WG, Goldberg AP. High-density-lipoprotein cholesterol in bodybuilders v powerlifters. Negative effects of androgen use. JAMA 1984; 252:507-513
  52. Kuipers H, Wijnen JA, Hartgens F, Willems SM. Influence of anabolic steroids on body composition, blood pressure, lipid profile and liver functions in body builders. Int J Sports Med 1991; 12:413-418
  53. Vermeulen A, Kaufman JM, Goemaere S, van Pottelberg I. Estradiol in elderly men. Aging Male 2002; 5:98-102
  54. van Velzen DM, Paldino A, Klaver M, Nota NM, Defreyne J, Hovingh GK, Thijs A, Simsek S, T'Sjoen G, den Heijer M. Cardiometabolic Effects of Testosterone in Transmen and Estrogen Plus Cyproterone Acetate in Transwomen. J Clin Endocrinol Metab 2019; 104:1937-1947
  55. Gava G, Mancini I, Cerpolini S, Baldassarre M, Seracchioli R, Meriggiola MC. Testosterone undecanoate and testosterone enanthate injections are both effective and safe in transmen over 5 years of administration. Clin Endocrinol (Oxf) 2018; 89:878-886
  56. Maraka S, Singh Ospina N, Rodriguez-Gutierrez R, Davidge-Pitts CJ, Nippoldt TB, Prokop LJ, Murad MH. Sex Steroids and Cardiovascular Outcomes in Transgender Individuals: A Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 2017; 102:3914-3923
  57. Leemaqz SY, Kyinn M, Banks K, Sarkodie E, Goldstein D, Irwig MS. Lipid profiles and hypertriglyceridemia among transgender and gender diverse adults on gender-affirming hormone therapy. J Clin Lipidol 2023; 17:103-111
  58. Allen AN, Jiao R, Day P, Pagels P, Gimpel N, SoRelle JA. Dynamic Impact of Hormone Therapy on Laboratory Values in Transgender Patients over Time. J Appl Lab Med 2021; 6:27-40
  59. Millington K, Chan YM. Lipoprotein subtypes after testosterone therapy in transmasculine adolescents. J Clin Lipidol 2021; 15:840-844
  60. Kaplan SA, Lin J, Johnson-Levonas AO, Shah AK, Meehan AG. Increased occurrence of marked elevations of lipoprotein(a) in ageing, hypercholesterolaemic men with low testosterone. Aging Male 2010; 13:40-43
  61. Henriksson P, Angelin B, Berglund L. Hormonal regulation of serum Lp (a) levels. Opposite effects after estrogen treatment and orchidectomy in males with prostatic carcinoma. J Clin Invest 1992; 89:1166-1171
  62. Zmunda JM, Thompson PD, Dickenson R, Bausserman LL. Testosterone decreases lipoprotein(a) in men. Am J Cardiol 1996; 77:1244-1247
  63. Marcovina SM, Lippi G, Bagatell CJ, Bremner WJ. Testosterone-induced suppression of lipoprotein(a) in normal men; relation to basal lipoprotein(a) level. Atherosclerosis 1996; 122:89-95
  64. Glazer G. Atherogenic effects of anabolic steroids on serum lipid levels. A literature review. Arch Intern Med1991; 151:1925-1933
  65. Langer C, Gansz B, Goepfert C, Engel T, Uehara Y, von Dehn G, Jansen H, Assmann G, von Eckardstein A. Testosterone up-regulates scavenger receptor BI and stimulates cholesterol efflux from macrophages. Biochem Biophys Res Commun 2002; 296:1051-1057
  66. Tan KC, Shiu SW, Kung AW. Alterations in hepatic lipase and lipoprotein subfractions with transdermal testosterone replacement therapy. Clin Endocrinol (Oxf) 1999; 51:765-769
  67. Zmuda JM, Fahrenbach MC, Younkin BT, Bausserman LL, Terry RB, Catlin DH, Thompson PD. The effect of testosterone aromatization on high-density lipoprotein cholesterol level and postheparin lipolytic activity. Metabolism 1993; 42:446-450
  68. Sorva R, Kuusi T, Taskinen MR, Perheentupa J, Nikkila EA. Testosterone substitution increases the activity of lipoprotein lipase and hepatic lipase in hypogonadal males. Atherosclerosis 1988; 69:191-197
  69. Haffner SM, Kushwaha RS, Foster DM, Applebaum-Bowden D, Hazzard WR. Studies on the metabolic mechanism of reduced high density lipoproteins during anabolic steroid therapy. Metabolism 1983; 32:413-420
  70. Croston GE, Milan LB, Marschke KB, Reichman M, Briggs MR. Androgen receptor-mediated antagonism of estrogen-dependent low density lipoprotein receptor transcription in cultured hepatocytes. Endocrinology 1997; 138:3779-3786
  71. Yeap BB, Dwivedi G. Androgens and Cardiovascular Disease in Men. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  72. Zhao J, Zhu S, Sun L, Meng F, Zhao L, Zhao Y, Tian H, Li P, Niu Y. Androgen deprivation therapy for prostate cancer is associated with cardiovascular morbidity and mortality: a meta-analysis of population-based observational studies. PLoS One 2014; 9:e107516
  73. Carneiro A, Sasse AD, Wagner AA, Peixoto G, Kataguiri A, Neto AS, Bianco BA, Chang P, Pompeo AC, Tobias-Machado M. Cardiovascular events associated with androgen deprivation therapy in patients with prostate cancer: a systematic review and meta-analysis. World J Urol 2015; 33:1281-1289
  74. Nguyen PL, Je Y, Schutz FA, Hoffman KE, Hu JC, Parekh A, Beckman JA, Choueiri TK. Association of androgen deprivation therapy with cardiovascular death in patients with prostate cancer: a meta-analysis of randomized trials. JAMA 2011; 306:2359-2366
  75. Bosco C, Bosnyak Z, Malmberg A, Adolfsson J, Keating NL, Van Hemelrijck M. Quantifying observational evidence for risk of fatal and nonfatal cardiovascular disease following androgen deprivation therapy for prostate cancer: a meta-analysis. Eur Urol 2015; 68:386-396
  76. Layton JB, Meier CR, Sharpless JL, Sturmer T, Jick SS, Brookhart MA. Comparative Safety of Testosterone Dosage Forms. JAMA Intern Med 2015; 175:1187-1196
  77. Basaria S, Coviello AD, Travison TG, Storer TW, Farwell WR, Jette AM, Eder R, Tennstedt S, Ulloor J, Zhang A, Choong K, Lakshman KM, Mazer NA, Miciek R, Krasnoff J, Elmi A, Knapp PE, Brooks B, Appleman E, Aggarwal S, Bhasin G, Hede-Brierley L, Bhatia A, Collins L, LeBrasseur N, Fiore LD, Bhasin S. Adverse events associated with testosterone administration. N Engl J Med 2010; 363:109-122
  78. Srinivas-Shankar U, Roberts SA, Connolly MJ, O'Connell MD, Adams JE, Oldham JA, Wu FC. Effects of testosterone on muscle strength, physical function, body composition, and quality of life in intermediate-frail and frail elderly men: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2010; 95:639-650
  79. Basaria S, Harman SM, Travison TG, Hodis H, Tsitouras P, Budoff M, Pencina KM, Vita J, Dzekov C, Mazer NA, Coviello AD, Knapp PE, Hally K, Pinjic E, Yan M, Storer TW, Bhasin S. Effects of Testosterone Administration for 3 Years on Subclinical Atherosclerosis Progression in Older Men With Low or Low-Normal Testosterone Levels: A Randomized Clinical Trial. JAMA 2015; 314:570-581
  80. Budoff MJ, Ellenberg SS, Lewis CE, Mohler ER, 3rd, Wenger NK, Bhasin S, Barrett-Connor E, Swerdloff RS, Stephens-Shields A, Cauley JA, Crandall JP, Cunningham GR, Ensrud KE, Gill TM, Matsumoto AM, Molitch ME, Nakanishi R, Nezarat N, Matsumoto S, Hou X, Basaria S, Diem SJ, Wang C, Cifelli D, Snyder PJ. Testosterone Treatment and Coronary Artery Plaque Volume in Older Men With Low Testosterone. JAMA2017; 317:708-716
  81. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med 2013; 11:108
  82. Borst SE, Shuster JJ, Zou B, Ye F, Jia H, Wokhlu A, Yarrow JF. Cardiovascular risks and elevation of serum DHT vary by route of testosterone administration: a systematic review and meta-analysis. BMC Med 2014; 12:211
  83. Calof OM, Singh AB, Lee ML, Kenny AM, Urban RJ, Tenover JL, Bhasin S. Adverse events associated with testosterone replacement in middle-aged and older men: a meta-analysis of randomized, placebo-controlled trials. J Gerontol A Biol Sci Med Sci 2005; 60:1451-1457
  84. Corona G, Maseroli E, Rastrelli G, Isidori AM, Sforza A, Mannucci E, Maggi M. Cardiovascular risk associated with testosterone-boosting medications: a systematic review and meta-analysis. Expert Opin Drug Saf 2014; 13:1327-1351
  85. Ruige JB, Ouwens DM, Kaufman JM. Beneficial and adverse effects of testosterone on the cardiovascular system in men. J Clin Endocrinol Metab 2013; 98:4300-4310
  86. Elliott J, Kelly SE, Millar AC, Peterson J, Chen L, Johnston A, Kotb A, Skidmore B, Bai Z, Mamdani M, Wells GA. Testosterone therapy in hypogonadal men: a systematic review and network meta-analysis. BMJ Open2017; 7:e015284
  87. Alexander GC, Iyer G, Lucas E, Lin D, Singh S. Cardiovascular Risks of Exogenous Testosterone Use Among Men: A Systematic Review and Meta-Analysis. Am J Med 2017; 130:293-305
  88. Hudson J, Cruickshank M, Quinton R, Aucott L, Aceves-Martins M, Gillies K, Bhasin S, Snyder PJ, Ellenberg SS, Grossmann M, Travison TG, Gianatti EJ, van der Schouw YT, Emmelot-Vonk MH, Giltay EJ, Hackett G, Ramachandran S, Svartberg J, Hildreth KL, Groti Antonic K, Brock GB, Tenover JL, Tan HM, Kong CHC, Tan WS, Marks LS, Ross RJ, Schwartz RS, Manson P, Roberts S, Andersen MS, Magnussen LV, Hernandez R, Oliver N, Wu F, Dhillo WS, Bhattacharya S, Brazzelli M, Jayasena CN. Adverse cardiovascular events and mortality in men during testosterone treatment: an individual patient and aggregate data meta-analysis. Lancet Healthy Longev 2022; 3:e381-e393
  89. Marriott RJ, Harse J, Murray K, Yeap BB. Systematic review and meta-analyses on associations of endogenous testosterone concentration with health outcomes in community-dwelling men. BMJ Open 2021; 11:e048013
  90. Bhasin S, Lincoff AM, Basaria S, Bauer DC, Boden WE, Cunningham GR, Davey D, Dubcenco E, Fukumoto S, Garcia M, Granger CB, Kalahasti V, Khera M, Miller MG, Mitchell LM, O'Leary MP, Pencina KM, Snyder PJ, Thompson IM, Jr., Travison TG, Wolski K, Nissen SE, Investigators TS. Effects of long-term testosterone treatment on cardiovascular outcomes in men with hypogonadism: Rationale and design of the TRAVERSE study. Am Heart J 2022; 245:41-50
  91. Bittner V. Lipoprotein abnormalities related to women's health. Am J Cardiol 2002; 90:77i-84i
  92. Phan BA, Toth PP. Dyslipidemia in women: etiology and management. Int J Womens Health 2014; 6:185-194
  93. Wang X, Magkos F, Mittendorfer B. Sex differences in lipid and lipoprotein metabolism: it's not just about sex hormones. J Clin Endocrinol Metab 2011; 96:885-893
  94. Cifkova R, Krajcoviechova A. Dyslipidemia and cardiovascular disease in women. Curr Cardiol Rep 2015; 17:609
  95. Jenner JL, Ordovas JM, Lamon-Fava S, Schaefer MM, Wilson PW, Castelli WP, Schaefer EJ. Effects of age, sex, and menopausal status on plasma lipoprotein(a) levels. The Framingham Offspring Study. Circulation1993; 87:1135-1141
  96. Lip GY, Blann AD, Jones AF, Beevers DG. Effects of hormone-replacement therapy on hemostatic factors, lipid factors, and endothelial function in women undergoing surgical menopause: implications for prevention of atherosclerosis. Am Heart J 1997; 134:764-771
  97. Godsland IF. Effects of postmenopausal hormone replacement therapy on lipid, lipoprotein, and apolipoprotein (a) concentrations: analysis of studies published from 1974-2000. Fertil Steril 2001; 75:898-915
  98. Lobo RA. Clinical review 27: Effects of hormonal replacement on lipids and lipoproteins in postmenopausal women. J Clin Endocrinol Metab 1991; 73:925-930
  99. Chait A, Subramanian S, Brunzell JD. Genetic Disorders of Triglyceride Metabolism. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA)2000.
  100. Goodman MP. Are all estrogens created equal? A review of oral vs. transdermal therapy. J Womens Health (Larchmt) 2012; 21:161-169
  101. Smith GI, Reeds DN, Okunade AL, Patterson BW, Mittendorfer B. Systemic delivery of estradiol, but not testosterone or progesterone, alters very low density lipoprotein-triglyceride kinetics in postmenopausal women. J Clin Endocrinol Metab 2014; 99:E1306-1310
  102. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA 1995; 273:199-208
  103. Darling GM, Johns JA, McCloud PI, Davis SR. Estrogen and progestin compared with simvastatin for hypercholesterolemia in postmenopausal women. N Engl J Med 1997; 337:595-601
  104. Tsuda M, Sanada M, Nakagawa H, Kodama I, Sakashita T, Ohama K. Phenotype of apolipoprotein E influences the lipid metabolic response of postmenopausal women to hormone replacement therapy. Maturitas2001; 38:297-304
  105. Herrington DM, Howard TD, Hawkins GA, Reboussin DM, Xu J, Zheng SL, Brosnihan KB, Meyers DA, Bleecker ER. Estrogen-receptor polymorphisms and effects of estrogen replacement on high-density lipoprotein cholesterol in women with coronary disease. N Engl J Med 2002; 346:967-974
  106. Liebeskind A, Thompson J, Wilson D. Reproductive Health and its Impact on Lipid Management in Adolescent and Young Adult Females. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  107. Kim JJ, Choi YM. Dyslipidemia in women with polycystic ovary syndrome. Obstet Gynecol Sci 2013; 56:137-142
  108. Wild RA. Dyslipidemia in PCOS. Steroids 2012; 77:295-299
  109. Wild RA, Rizzo M, Clifton S, Carmina E. Lipid levels in polycystic ovary syndrome: systematic review and meta-analysis. Fertil Steril 2011; 95:1073-1079 e1071-1011
  110. Wang M, Zhao D, Xu L, Guo W, Nie L, Lei Y, Long Y, Liu M, Wang Y, Zhang X, Zhang L, Li H, Zhang J, Yuan D, Yue L. Role of PCSK9 in lipid metabolic disorders and ovarian dysfunction in polycystic ovary syndrome. Metabolism 2019; 94:47-58
  111. Vatannejad A, Salimi F, Moradi N, Fouani FZ, Zandieh Z, Ansaripour S, Sadeghi A, Fadaei R. Evaluation of angiopoietin-like protein 3 (ANGPTL3) levels in polycystic ovary syndrome. Life Sci 2020; 263:118595
  112. Brinton EA. Oral estrogen replacement therapy in postmenopausal women selectively raises levels and production rates of lipoprotein A-I and lowers hepatic lipase activity without lowering the fractional catabolic rate. Arterioscler Thromb Vasc Biol 1996; 16:431-440
  113. Hargrove GM, Junco A, Wong NC. Hormonal regulation of apolipoprotein AI. J Mol Endocrinol 1999; 22:103-111
  114. Lamon-Fava S, Postfai B, Diffenderfer M, DeLuca C, O'Connor J, Jr., Welty FK, Dolnikowski GG, Barrett PH, Schaefer EJ. Role of the estrogen and progestin in hormonal replacement therapy on apolipoprotein A-I kinetics in postmenopausal women. Arterioscler Thromb Vasc Biol 2006; 26:385-391
  115. Schaefer EJ, Foster DM, Zech LA, Lindgren FT, Brewer HB, Jr., Levy RI. The effects of estrogen administration on plasma lipoprotein metabolism in premenopausal females. J Clin Endocrinol Metab 1983; 57:262-267
  116. Walsh BW, Li H, Sacks FM. Effects of postmenopausal hormone replacement with oral and transdermal estrogen on high density lipoprotein metabolism. J Lipid Res 1994; 35:2083-2093
  117. Applebaum DM, Goldberg AP, Pykalisto OJ, Brunzell JD, Hazzard WR. Effect of estrogen on post-heparin lipolytic activity. Selective decline in hepatic triglyceride lipase. J Clin Invest 1977; 59:601-608
  118. Applebaum-Bowden D, McLean P, Steinmetz A, Fontana D, Matthys C, Warnick GR, Cheung M, Albers JJ, Hazzard WR. Lipoprotein, apolipoprotein, and lipolytic enzyme changes following estrogen administration in postmenopausal women. J Lipid Res 1989; 30:1895-1906
  119. Tikkanen MJ, Nikkila EA, Kuusi T, Sipinen SU. High density lipoprotein-2 and hepatic lipase: reciprocal changes produced by estrogen and norgestrel. J Clin Endocrinol Metab 1982; 54:1113-1117
  120. Lopez D, McLean MP. Estrogen regulation of the scavenger receptor class B gene: Anti-atherogenic or steroidogenic, is there a priority? Mol Cell Endocrinol 2006; 247:22-33
  121. Campos H, Walsh BW, Judge H, Sacks FM. Effect of estrogen on very low density lipoprotein and low density lipoprotein subclass metabolism in postmenopausal women. J Clin Endocrinol Metab 1997; 82:3955-3963
  122. Eriksson M, Berglund L, Rudling M, Henriksson P, Angelin B. Effects of estrogen on low density lipoprotein metabolism in males. Short-term and long-term studies during hormonal treatment of prostatic carcinoma. J Clin Invest 1989; 84:802-810
  123. Karjalainen A, Heikkinen J, Savolainen MJ, Backstrom AC, Kesaniemi YA. Mechanisms regulating LDL metabolism in subjects on peroral and transdermal estrogen replacement therapy. Arterioscler Thromb Vasc Biol 2000; 20:1101-1106
  124. Walsh BW, Schiff I, Rosner B, Greenberg L, Ravnikar V, Sacks FM. Effects of postmenopausal estrogen replacement on the concentrations and metabolism of plasma lipoproteins. N Engl J Med 1991; 325:1196-1204
  125. Parini P, Angelin B, Rudling M. Importance of estrogen receptors in hepatic LDL receptor regulation. Arterioscler Thromb Vasc Biol 1997; 17:1800-1805
  126. Semenkovich CF, Ostlund RE, Jr. Estrogens induce low-density lipoprotein receptor activity and decrease intracellular cholesterol in human hepatoma cell line Hep G2. Biochemistry 1987; 26:4987-4992
  127. Windler EE, Kovanen PT, Chao YS, Brown MS, Havel RJ, Goldstein JL. The estradiol-stimulated lipoprotein receptor of rat liver. A binding site that membrane mediates the uptake of rat lipoproteins containing apoproteins B and E. J Biol Chem 1980; 255:10464-10471
  128. Kovanen PT, Brown MS, Goldstein JL. Increased binding of low density lipoprotein to liver membranes from rats treated with 17 alpha-ethinyl estradiol. J Biol Chem 1979; 254:11367-11373
  129. Persson L, Henriksson P, Westerlund E, Hovatta O, Angelin B, Rudling M. Endogenous estrogens lower plasma PCSK9 and LDL cholesterol but not Lp(a) or bile acid synthesis in women. Arterioscler Thromb Vasc Biol 2012; 32:810-814
  130. Persson L, Galman C, Angelin B, Rudling M. Importance of proprotein convertase subtilisin/kexin type 9 in the hormonal and dietary regulation of rat liver low-density lipoprotein receptors. Endocrinology 2009; 150:1140-1146
  131. Ghosh M, Galman C, Rudling M, Angelin B. Influence of physiological changes in endogenous estrogen on circulating PCSK9 and LDL cholesterol. J Lipid Res 2015; 56:463-469
  132. Glueck CJ, Fallat RW, Scheel D. Effects of estrogenic compounds on triglyceride kinetics. Metabolism 1975; 24:537-545
  133. Kissebah AH, Harrigan P, Wynn V. Mechanism of hypertriglyceridaemia associated with contraceptive steroids. Horm Metab Res 1973; 5:184-190
  134. Tikkanen MJ, Kuusi T, Nikkila EA, Sane T. Very low density lipoprotein triglyceride kinetics during hepatic lipase suppression by estrogen. Studies on the physiological role of hepatic endothelial lipase. FEBS Lett1985; 181:160-164
  135. Sitruk-Ware R, Nath A. Characteristics and metabolic effects of estrogen and progestins contained in oral contraceptive pills. Best Pract Res Clin Endocrinol Metab 2013; 27:13-24
  136. Johansson S, Vedin A, Wilhelmsson C. Myocardial infarction in women. Epidemiol Rev 1983; 5:67-95
  137. Lerner DJ, Kannel WB. Patterns of coronary heart disease morbidity and mortality in the sexes: a 26-year follow-up of the Framingham population. Am Heart J 1986; 111:383-390
  138. Yusuf S, Hawken S, Ounpuu S, Dans T, Avezum A, Lanas F, McQueen M, Budaj A, Pais P, Varigos J, Lisheng L, Investigators IS. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet 2004; 364:937-952
  139. Faubion SS, Kuhle CL, Shuster LT, Rocca WA. Long-term health consequences of premature or early menopause and considerations for management. Climacteric 2015; 18:483-491
  140. Gordon T, Kannel WB, Hjortland MC, McNamara PM. Menopause and coronary heart disease. The Framingham Study. Ann Intern Med 1978; 89:157-161
  141. Kannel WB, Hjortland MC, McNamara PM, Gordon T. Menopause and risk of cardiovascular disease: the Framingham study. Ann Intern Med 1976; 85:447-452
  142. Atsma F, Bartelink ML, Grobbee DE, van der Schouw YT. Postmenopausal status and early menopause as independent risk factors for cardiovascular disease: a meta-analysis. Menopause 2006; 13:265-279
  143. Zhu D, Chung HF, Dobson AJ, Pandeya N, Giles GG, Bruinsma F, Brunner EJ, Kuh D, Hardy R, Avis NE, Gold EB, Derby CA, Matthews KA, Cade JE, Greenwood DC, Demakakos P, Brown DE, Sievert LL, Anderson D, Hayashi K, Lee JS, Mizunuma H, Tillin T, Simonsen MK, Adami HO, Weiderpass E, Mishra GD. Age at natural menopause and risk of incident cardiovascular disease: a pooled analysis of individual patient data. Lancet Public Health 2019; 4:e553-e564
  144. Lubiszewska B, Kruk M, Broda G, Ksiezycka E, Piotrowski W, Kurjata P, Zielinski T, Ploski R. The impact of early menopause on risk of coronary artery disease (PREmature Coronary Artery Disease In Women--PRECADIW case-control study). Eur J Prev Cardiol 2012; 19:95-101
  145. Muka T, Oliver-Williams C, Kunutsor S, Laven JS, Fauser BC, Chowdhury R, Kavousi M, Franco OH. Association of Age at Onset of Menopause and Time Since Onset of Menopause With Cardiovascular Outcomes, Intermediate Vascular Traits, and All-Cause Mortality: A Systematic Review and Meta-analysis. JAMA Cardiol 2016; 1:767-776
  146. Wellons M, Ouyang P, Schreiner PJ, Herrington DM, Vaidya D. Early menopause predicts future coronary heart disease and stroke: the Multi-Ethnic Study of Atherosclerosis. Menopause 2012; 19:1081-1087
  147. Barrett-Connor E, Bush TL. Estrogen and coronary heart disease in women. JAMA 1991; 265:1861-1867
  148. Grodstein F, Manson JE, Colditz GA, Willett WC, Speizer FE, Stampfer MJ. A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease. Ann Intern Med 2000; 133:933-941
  149. Grodstein F, Stampfer MJ, Manson JE, Colditz GA, Willett WC, Rosner B, Speizer FE, Hennekens CH. Postmenopausal estrogen and progestin use and the risk of cardiovascular disease. N Engl J Med 1996; 335:453-461
  150. Henderson BE, Paganini-Hill A, Ross RK. Decreased mortality in users of estrogen replacement therapy. Arch Intern Med 1991; 151:75-78
  151. Psaty BM, Heckbert SR, Atkins D, Lemaitre R, Koepsell TD, Wahl PW, Siscovick DS, Wagner EH. The risk of myocardial infarction associated with the combined use of estrogens and progestins in postmenopausal women. Arch Intern Med 1994; 154:1333-1339
  152. Sidney S, Petitti DB, Quesenberry CP, Jr. Myocardial infarction and the use of estrogen and estrogen-progestogen in postmenopausal women. Ann Intern Med 1997; 127:501-508
  153. Stampfer MJ, Colditz GA, Willett WC, Manson JE, Rosner B, Speizer FE, Hennekens CH. Postmenopausal estrogen therapy and cardiovascular disease. Ten-year follow-up from the nurses' health study. N Engl J Med1991; 325:756-762
  154. Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, Vittinghoff E. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA 1998; 280:605-613
  155. Grady D, Herrington D, Bittner V, Blumenthal R, Davidson M, Hlatky M, Hsia J, Hulley S, Herd A, Khan S, Newby LK, Waters D, Vittinghoff E, Wenger N, Group HR. Cardiovascular disease outcomes during 6.8 years of hormone therapy: Heart and Estrogen/progestin Replacement Study follow-up (HERS II). JAMA 2002; 288:49-57
  156. Shlipak MG, Simon JA, Vittinghoff E, Lin F, Barrett-Connor E, Knopp RH, Levy RI, Hulley SB. Estrogen and progestin, lipoprotein(a), and the risk of recurrent coronary heart disease events after menopause. JAMA2000; 283:1845-1852
  157. Manson JE, Hsia J, Johnson KC, Rossouw JE, Assaf AR, Lasser NL, Trevisan M, Black HR, Heckbert SR, Detrano R, Strickland OL, Wong ND, Crouse JR, Stein E, Cushman M, Women's Health Initiative I. Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med 2003; 349:523-534
  158. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R, Brzyski R, Caan B, Chlebowski R, Curb D, Gass M, Hays J, Heiss G, Hendrix S, Howard BV, Hsia J, Hubbell A, Jackson R, Johnson KC, Judd H, Kotchen JM, Kuller L, LaCroix AZ, Lane D, Langer RD, Lasser N, Lewis CE, Manson J, Margolis K, Ockene J, O'Sullivan MJ, Phillips L, Prentice RL, Ritenbaugh C, Robbins J, Rossouw JE, Sarto G, Stefanick ML, Van Horn L, Wactawski-Wende J, Wallace R, Wassertheil-Smoller S, Women's Health Initiative Steering C. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 2004; 291:1701-1712
  159. Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE, Prentice RL, Anderson G, Howard BV, Thomson CA, LaCroix AZ, Wactawski-Wende J, Jackson RD, Limacher M, Margolis KL, Wassertheil-Smoller S, Beresford SA, Cauley JA, Eaton CB, Gass M, Hsia J, Johnson KC, Kooperberg C, Kuller LH, Lewis CE, Liu S, Martin LW, Ockene JK, O'Sullivan MJ, Powell LH, Simon MS, Van Horn L, Vitolins MZ, Wallace RB. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA 2013; 310:1353-1368
  160. Harman SM, Vittinghoff E, Brinton EA, Budoff MJ, Cedars MI, Lobo RA, Merriam GR, Miller VM, Naftolin F, Pal L, Santoro N, Taylor HS, Black DM. Timing and duration of menopausal hormone treatment may affect cardiovascular outcomes. Am J Med 2011; 124:199-205
  161. Rossouw JE, Prentice RL, Manson JE, Wu L, Barad D, Barnabei VM, Ko M, LaCroix AZ, Margolis KL, Stefanick ML. Postmenopausal hormone therapy and risk of cardiovascular disease by age and years since menopause. JAMA 2007; 297:1465-1477
  162. Salpeter SR, Walsh JM, Greyber E, Salpeter EE. Brief report: Coronary heart disease events associated with hormone therapy in younger and older women. A meta-analysis. J Gen Intern Med 2006; 21:363-366
  163. Boardman HM, Hartley L, Eisinga A, Main C, Roque i Figuls M, Bonfill Cosp X, Gabriel Sanchez R, Knight B. Hormone therapy for preventing cardiovascular disease in post-menopausal women. Cochrane Database Syst Rev 2015:CD002229
  164. Schierbeck LL, Rejnmark L, Tofteng CL, Stilgren L, Eiken P, Mosekilde L, Kober L, Jensen JE. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ 2012; 345:e6409
  165. Bassuk SS, Manson JE. The timing hypothesis: Do coronary risks of menopausal hormone therapy vary by age or time since menopause onset? Metabolism 2016; 65:794-803
  166. Bray PF, Larson JC, Lacroix AZ, Manson J, Limacher MC, Rossouw JE, Lasser NL, Lawson WE, Stefanick ML, Langer RD, Margolis KL, Women's Health Initiative I. Usefulness of baseline lipids and C-reactive protein in women receiving menopausal hormone therapy as predictors of treatment-related coronary events. Am J Cardiol 2008; 101:1599-1605
  167. Wild RA, Wu C, Curb JD, Martin LW, Phillips L, Stefanick M, Trevisan M, Manson JE. Coronary heart disease events in the Women's Health Initiative hormone trials: effect modification by metabolic syndrome: a nested case-control study within the Women's Health Initiative randomized clinical trials. Menopause 2013; 20:254-260
  168. Wild RA, Hovey KM, Andrews C, Robinson JG, Kaunitz AM, Manson JE, Crandall CJ, Paragallo R, Shufelt C, Merz CNB. Cardiovascular disease (CVD) risk scores, age, or years since menopause to predict cardiovascular disease in the Women's Health Initiative. Menopause 2021; 28:610-618
  169. Toh S, Hernandez-Diaz S, Logan R, Rossouw JE, Hernan MA. Coronary heart disease in postmenopausal recipients of estrogen plus progestin therapy: does the increased risk ever disappear? A randomized trial. Ann Intern Med 2010; 152:211-217
  170. Hernan MA, Alonso A, Logan R, Grodstein F, Michels KB, Willett WC, Manson JE, Robins JM. Observational studies analyzed like randomized experiments: an application to postmenopausal hormone therapy and coronary heart disease. Epidemiology 2008; 19:766-779
  171. Herrington DM, Reboussin DM, Brosnihan KB, Sharp PC, Shumaker SA, Snyder TE, Furberg CD, Kowalchuk GJ, Stuckey TD, Rogers WJ, Givens DH, Waters D. Effects of estrogen replacement on the progression of coronary-artery atherosclerosis. N Engl J Med 2000; 343:522-529
  172. Hodis HN, Mack WJ, Azen SP, Lobo RA, Shoupe D, Mahrer PR, Faxon DP, Cashin-Hemphill L, Sanmarco ME, French WJ, Shook TL, Gaarder TD, Mehra AO, Rabbani R, Sevanian A, Shil AB, Torres M, Vogelbach KH, Selzer RH, Women's Estrogen-Progestin Lipid-Lowering Hormone Atherosclerosis Regression Trial Research G. Hormone therapy and the progression of coronary-artery atherosclerosis in postmenopausal women. N Engl J Med 2003; 349:535-545
  173. Waters DD, Alderman EL, Hsia J, Howard BV, Cobb FR, Rogers WJ, Ouyang P, Thompson P, Tardif JC, Higginson L, Bittner V, Steffes M, Gordon DJ, Proschan M, Younes N, Verter JI. Effects of hormone replacement therapy and antioxidant vitamin supplements on coronary atherosclerosis in postmenopausal women: a randomized controlled trial. JAMA 2002; 288:2432-2440
  174. Angerer P, Stork S, Kothny W, Schmitt P, von Schacky C. Effect of oral postmenopausal hormone replacement on progression of atherosclerosis : a randomized, controlled trial. Arterioscler Thromb Vasc Biol 2001; 21:262-268
  175. Manson JE, Allison MA, Rossouw JE, Carr JJ, Langer RD, Hsia J, Kuller LH, Cochrane BB, Hunt JR, Ludlam SE, Pettinger MB, Gass M, Margolis KL, Nathan L, Ockene JK, Prentice RL, Robbins J, Stefanick ML, Whi, Investigators W-C. Estrogen therapy and coronary-artery calcification. N Engl J Med 2007; 356:2591-2602
  176. Hodis HN, Mack WJ, Lobo RA, Shoupe D, Sevanian A, Mahrer PR, Selzer RH, Liu Cr CR, Liu Ch CH, Azen SP, Estrogen in the Prevention of Atherosclerosis Trial Research G. Estrogen in the prevention of atherosclerosis. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 2001; 135:939-953
  177. Hodis HN, Mack WJ, Henderson VW, Shoupe D, Budoff MJ, Hwang-Levine J, Li Y, Feng M, Dustin L, Kono N, Stanczyk FZ, Selzer RH, Azen SP, Group ER. Vascular Effects of Early versus Late Postmenopausal Treatment with Estradiol. N Engl J Med 2016; 374:1221-1231
  178. Roach RE, Helmerhorst FM, Lijfering WM, Stijnen T, Algra A, Dekkers OM. Combined oral contraceptives: the risk of myocardial infarction and ischemic stroke. Cochrane Database Syst Rev 2015:CD011054
  179. Chakhtoura Z, Canonico M, Gompel A, Scarabin PY, Plu-Bureau G. Progestogen-only contraceptives and the risk of acute myocardial infarction: a meta-analysis. J Clin Endocrinol Metab 2011; 96:1169-1174
  180. Peragallo Urrutia R, Coeytaux RR, McBroom AJ, Gierisch JM, Havrilesky LJ, Moorman PG, Lowery WJ, Dinan M, Hasselblad V, Sanders GD, Myers ER. Risk of acute thromboembolic events with oral contraceptive use: a systematic review and meta-analysis. Obstet Gynecol 2013; 122:380-389
  181. Baillargeon JP, McClish DK, Essah PA, Nestler JE. Association between the current use of low-dose oral contraceptives and cardiovascular arterial disease: a meta-analysis. J Clin Endocrinol Metab 2005; 90:3863-3870
  182. Khader YS, Rice J, John L, Abueita O. Oral contraceptives use and the risk of myocardial infarction: a meta-analysis. Contraception 2003; 68:11-17
  183. Zhao L, Zhu Z, Lou H, Zhu G, Huang W, Zhang S, Liu F. Polycystic ovary syndrome (PCOS) and the risk of coronary heart disease (CHD): a meta-analysis. Oncotarget 2016;
  184. de Groot PC, Dekkers OM, Romijn JA, Dieben SW, Helmerhorst FM. PCOS, coronary heart disease, stroke and the influence of obesity: a systematic review and meta-analysis. Hum Reprod Update 2011; 17:495-500
  185. Iftikhar S, Collazo-Clavell ML, Roger VL, St Sauver J, Brown RD, Jr., Cha S, Rhodes DJ. Risk of cardiovascular events in patients with polycystic ovary syndrome. Neth J Med 2012; 70:74-80
  186. Jacewicz-Swiecka M, Kowalska I. Polycystic ovary syndrome and the risk of cardiometabolic complications in longitudinal studies. Diabetes Metab Res Rev 2018; 34:e3054
  187. Guan C, Zahid S, Minhas AS, Ouyang P, Vaught A, Baker VL, Michos ED. Polycystic ovary syndrome: a "risk-enhancing" factor for cardiovascular disease. Fertil Steril 2022; 117:924-935
  188. Carmina E, Lobo RA. Is There Really Increased Cardiovascular Morbidity in Women with Polycystic Ovary Syndrome? J Womens Health (Larchmt) 2018; 27:1385-1388
  189. Meyer ML, Malek AM, Wild RA, Korytkowski MT, Talbott EO. Carotid artery intima-media thickness in polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod Update 2012; 18:112-126
  190. Gomez JMD, VanHise K, Stachenfeld N, Chan JL, Merz NB, Shufelt C. Subclinical cardiovascular disease and polycystic ovary syndrome. Fertil Steril 2022; 117:912-923
  191. Talbott EO, Zborowski JV, Rager JR, Boudreaux MY, Edmundowicz DA, Guzick DS. Evidence for an association between metabolic cardiovascular syndrome and coronary and aortic calcification among women with polycystic ovary syndrome. J Clin Endocrinol Metab 2004; 89:5454-5461
  192. Talbott EO, Zborowski J, Rager J, Stragand JR. Is there an independent effect of polycystic ovary syndrome (PCOS) and menopause on the prevalence of subclinical atherosclerosis in middle aged women? Vasc Health Risk Manag 2008; 4:453-462
  193. Shroff R, Kerchner A, Maifeld M, Van Beek EJ, Jagasia D, Dokras A. Young obese women with polycystic ovary syndrome have evidence of early coronary atherosclerosis. J Clin Endocrinol Metab 2007; 92:4609-4614
  194. Chang AY, Ayers C, Minhajuddin A, Jain T, Nurenberg P, de Lemos JA, Wild RA, Auchus RJ. Polycystic ovarian syndrome and subclinical atherosclerosis among women of reproductive age in the Dallas heart study. Clin Endocrinol (Oxf) 2011; 74:89-96
  195. Christian RC, Dumesic DA, Behrenbeck T, Oberg AL, Sheedy PF, 2nd, Fitzpatrick LA. Prevalence and predictors of coronary artery calcification in women with polycystic ovary syndrome. J Clin Endocrinol Metab2003; 88:2562-2568

 

Vasoactive Intestinal Peptide-Secreting Tumor (VIPoma)

ABSTRACT

A VIPoma is a neuroendocrine neoplasm secreting vasoactive intestinal peptide (VIP), usually presenting with severe watery secretory diarrhea, which can result in hypokalemia and metabolic acidosis and with flushes. Hypochlorhydria, stimulation of glycogenolysis, and hypercalcemia can be also found in VIPoma patients. Plasma VIP levels are elevated in all patients with the VIPoma syndrome, which is also known as “watery diarrhea, hypokalemia, achlorhydria (WDHA)-syndrome”, or “Verner-Morrison syndrome”. The majority of VIPomas are located in the pancreas (75%) and (usually young) patients can present with VIP-producing neuroblastoma, ganglioneuroblastoma, ganglioneuroma, pheochromocytoma and paraganglioma, or neoplasms of the retroperitoneum and mediastinum. The first treatment aim of a VIPoma patient is to correct the fluid and electrolyte deficits. Administration of a somatostatin analog (SSA) can decrease flushing and diarrhea, further aiding in the restoration of fluid and electrolyte imbalances. Surgical resection should be considered in patients with a locoregionally confined VIPoma. In patients with a metastatic or unresectable VIPoma, SSAs likely prolong progression-free survival. Other treatment options include peptide receptor radionuclide therapy (PRRT) with radiolabeled SSAs, interferon alpha, everolimus, sunitinib, cytotoxic chemotherapy, or liver-directed therapies.

 

INTRODUCTION

Vasoactive intestinal polypeptide (VIP) is a neurotransmitter found in the central nervous system, in neurons in the intestine, lungs, adrenals, pancreas and liver and in neuroendocrine cells in the pancreas (1). In the gastrointestinal tract, VIP stimulates contraction of enteric smooth muscle cells, secretion from the exocrine pancreas, gastrointestinal blood flow, and inhibits gastric acid secretion (2-4). A VIPoma is a neuroendocrine neoplasm (NEN) secreting VIP. VIP hypersecretion causes severe watery secretory diarrhea, which can result in hypokalemia and metabolic acidosis (VIPoma syndrome) (5).

 

HISTORY

In 1958 the US physician John V. Verner Jr. (1927-2022) and the Irish-US pathologist Ashton B. Morrison (1922-2008) reported on two patients with a VIPoma syndrome (6). Both patients presented with watery diarrhea and severe refractory hypokalemia and subsequently died of cardiac arrhythmias. Autopsy revealed pancreatic “islet cell” tumors in both patients (6). One of their patients was a 19-year-old male who also developed hypercalcemia and at autopsy hyperplasia of one of the parathyroid glands was found. The pituitary was not examined (6, 7). The publication by Verner and Morrison further cites 7 similar cases already published in the literature at that time (6) Thereafter, the VIPoma syndrome was also named “watery diarrhea, hypokalemia, achlorhydria (WDHA)-syndrome”, or “Verner-Morrison syndrome”. In the late 1960s and early 1970s, VIP was first isolated from the lungs and small intestine of experimental animals by the group of the Estonian scientist Viktor Mutt (1923-1998) in Sweden (8-10). In 1973, a radioimmunoassay for VIP became available and subsequently the British physician Stephen R. Bloom and colleagues could for the first time measure elevated VIP levels in the blood of a patient with the VIPoma syndrome (11). In 1983, the US gastroenterologist Mary G. Kane and colleagues injected five healthy subjects with porcine VIP, which resulted within 4 hours in high plasma VIP levels and was followed by secretory diarrhea in all patients (12).

 

CLINICAL PRESENTATION

Secretory diarrhea is the most characteristic symptom of a VIPoma. In severe cases, patients can produce up to 6-8L of watery stools per day. The stool is rich in electrolytes like potassium and bicarbonate, resulting in hypokalemia and metabolic acidosis in the VIPoma patient (13, 14). Another VIPoma symptom is facial flushing (occurring in 15-30% of patients). Hypochlorhydria, stimulation of glycogenolysis, and hypercalcemia can be diagnosed in patients with a VIPoma (5, 14-19). VIP has a structural homology with secretin, glucagon, and GIP which may account for the enhanced secretion of pancreatic enzymes, inhibition of gastric acid secretion, and glycogenolysis (9). The cause of the patchy erythematous flushing is not clear, but the flushing has been attributed to VIP, or to prostaglandins co-secreted by the tumor. Approximately 50% of patients have hypercalcemia, but again the mechanism of action is unknown. Hypercalcemia might be related to the co-secretion of parathyroid hormone related peptide (PTHrp) by the tumor (20, 21), or in specific cases coexisting primary hyperparathyroidism in the spectrum of the multiple endocrine neoplasia 1 (MEN1) syndrome (7).

Pancreatic VIPomas account for only 0.6–1.5% of all pancreatic neuroendocrine neoplasms (panNENs) (17) and approximately 2–6% of all functioning panNENs (17). The incidence is 0.05–0.2 cases per 1 million person-years with no gender predilection (15, 17, 18, 22). The mean age of these patients is 50.5 years (17). Pancreatic VIPomas can be associated with the MEN1 syndrome, but they are present in less than 1% of MEN1 patients (7, 23, 24). Around 75-90% of WDHA syndrome originates from a VIP-secreting panNEN. Approximately 70% of these pancreatic VIPomas are located in the body or tail and 30% in the head (18, 19, 25, 26). 10-25% of the WDHA syndrome derives from extra-pancreatic sources and can be found in patients with neuroblastoma, ganglioneuroblastoma, ganglioneuroma, pheochromocytoma and paraganglioma, and neoplasms of the retroperitoneum and mediastinum (5, 19, 27-30). The neurogenic tumors are more commonly found in the pediatric population (mean age 7.3 years). VIP-production from medullary thyroid carcinoma and lung neoplasms can also occur but this generally does not lead to the VIPoma / WDHA syndrome (31-33).

 

DIAGNOSIS

In the circulation, VIP has a very short half-life of less than 1 minute and, normally, plasma levels of VIP are low (below 20 pmol/L = 70 pg/mL) (34, 35). In the absence of a VIPoma, plasma VIP levels reflect the overflow of VIP from VIP-containing vascular nerves. By definition, plasma VIP levels should be elevated in all patients with the VIPoma syndrome. Bloom and colleagues measured plasma VIP levels in nearly 1000 patients with diarrhea and the diagnosis of VIPoma could be confirmed in all patients with plasma VIP levels greater than 60 pmol/L (= 203 pg/mL) (13, 34). In another series of 52 pancreatic VIPoma patients, elevated VIP levels were also measured with a median of 188 pmol/L (= 630 pg/mL - range 30-2131 pmol/L) (14). Moderately elevated plasma VIP levels can also be caused by gastrointestinal ischemia, renal insufficiency, or congestive heart failure (36-38).

The diameter of the primary pancreatic VIPoma is on average larger than 2 cm in 80% of patients (19). Therefore, these tumors can be easily detected with abdominal MRI, 3 phase CT, or endoscopic ultrasound (EUS). Additionally, a positron emission tomography (PET)-CT/MRI with 68Ga-labelled somatostatin analogs (DOTATATE, DOTANOC, DOTATOC) should be performed to determine, or exclude metastatic spread. In most centers, somatostatin receptor scintigraphy and SPECT using 111In-pentetreotide (OctreoScan) has become obsolete. In a small case series, 111In-pentetreotide scintigraphy proved to be superior to conventional radiological imaging for localizing the VIPoma and its metastases (39).

Similar to work-up for all NENs, a biopsy of the primary tumor or its metastases is recommended to confirm the diagnosis and for grading (Ki67 index), since the tumor grade can influence treatment decisions (17). An overview of the current panNEN staging and grading systems is provided in the chapter “Insulinoma” (40). Pancreatic VIPoma tumor cells usually express neuroendocrine differentiation markers (chromogranin-A, synaptophysin, INSM1), keratins, transcription factors, and somatostatin receptor subtype 2 (17). The extent of VIP expression can be variable given the rapid turnover of the protein synthesis. Secondary, or metachronous insulin secretion and/or positive insulin immunohistochemistry on the tumor specimen is generally associated with poor survival (41-43).

In patients with metastatic VIPoma, the 5-years survival is 60% (14, 16). Patients with high circulating VIP levels (plasma VIP ≥ 5xULN) have a poorer prognosis than those with moderately elevated levels (plasma VIP <5xULN) (16).

 

TREATMENT

Correction of Fluid and Electrolyte Deficits

The first treatment aim in a patient with a VIPoma is to correct the fluid and electrolyte deficits. In the majority of severe cases, intravenous resuscitation with saline, potassium and bicarbonate is required. Administration of a somatostatin analog (SSA) can decrease the secretory diarrhea, further aiding in the restoration of fluid and electrolyte imbalances (13, 44, 45). In the acute setting, the SSA octreotide can be administered subcutaneously, or via continuous intravenous infusion (46).

Surgery

After initial stabilization, a surgical resection should be performed in patients with a locoregionally confined VIPoma. The 5-year overall survival after surgery of patients with a localized VIPoma is >90% (14, 16, 19). In these patients the symptomatology of the VIPoma syndrome also completely resolved after surgery (16). Extended surgical resection, also involving the liver, can be considered in selected patients with limited liver metastases (47).

In case of an unresectable VIPoma, treatment is focused on tumor stabilization and control of VIP hypersecretion and symptoms (16). In general, anti-tumor therapy is similar to that used for other non-functioning and functioning panNENs and described in the guidelines by ENETS, NANETS and ESMO (48-50).

Somatostatin Analogs

Somatostatin analogs (SSAs) represent the first-line palliative treatment for metastatic or unresectable VIPomas. SSAs can have an antiproliferative effect, based on randomized trials with low grade (G1-G2) panNEN. In the CLARINET trial, including grade 1-2 panNENs, treatment with lanreotide autogel (120 mg every 4 weeks) prolonged median progression-free survival (PFS) from 18 to 38 months as compared to placebo by slowing tumor growth (51, 52). Treatment with SSAs results in a reduction of diarrhea episodes and volume in approximately 65-85% of VIPoma patients (15, 16, 45, 53, 54). It is, therefore, recommended to continue SSAs for symptom control when further lines of treatment are instituted for the control of tumor progression.

Everolimus

Everolimus is registered for the second-line treatment of G1-2 panNENs based on the result of the RADIANT-3 trial. In this study, 24% of patients had a functioning (= hormone-secreting) panNEN and treatment with everolimus (10 mg / day) improved median progression-free survival by 6.4 months compared with placebo. Everolimus treatment was associated with a (statistically not significant) overall survival benefit of 6.3 months (55, 56). Only a few VIPoma patients treated with Everolimus have been reported. In these patients, a symptomatic response was found in less than 10% of patients (15).

Sunitinib

In a randomized controlled trial in patients with G1-2 panNENs, second-line sunitinib treatment (37.5 mg/day) resulted in an increased progression-free survival by 5.9 months compared to placebo (57, 58). Two patients with a VIPoma were included in this trial, but they were both treated with placebo (57). In case series, a symptomatic response rate of 30-100% has been described for VIPoma patients treated with sunitinib (15, 16, 59, 60).

Other Medical Options

Next to SSAs, interferon-alpha is an established first-line antiproliferative and anti-secretory therapy for NENs of the gastrointestinal tract and pancreas either as monotherapy, or in combination with an SSA. However, the many side-effects mainly preclude its widespread use. Variable symptomatic responses with this therapy in VIPoma patients have been reported (61, 62). Prednisone has also been occasionally used to control the diarrhea frequency and stool volume in selected cases (45, 63).

Peptide Receptor Radionuclide Therapy

Peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE results in a response rate of 55% for panNENs, with a median PFS of 30 months and median overall survival (OS) of 71 months (64). PRRT with 177Lu-DOTATATE has only been reported in a limited number of patients with a VIPoma. In case series, the symptomatic response rate of VIPomas to this therapy was approximately 80% and disease control rate was 67% (15, 65, 66). Withdrawal from non-radioactive SSAs can lead to swift recurrence of severe watery diarrhea, providing rationale to limit the time for SSA withdrawal before PRRT cycles with 177Lu-DOTATATE to a very minimum e.g., by continued use of short-acting octreotide until shortly before the administration of this therapy (64).

Liver Directed Therapy

In patients with liver-dominant disease, liver metastases can be resected or treated by bland embolization, radioembolization (SIRT), radiofrequency ablation (RFA), microwave and cryoablation, high-intensity focused ultrasound (HIFU), laser, brachytherapy and irreversible electroporation (IRE) depending on local availability (47). Reduction of liver tumor burden was associated with a symptomatic response of VIPomas in small series (15, 16, 67). Orthotopic liver transplantation with removal of the diseased liver in VIPoma patients preoperatively diagnosed with “liver-only” disease can result in an improved disease course, or even complete cure (68-70).

Chemotherapy

Chemotherapy is also effective for the treatment of panNEN with symptomatic and tumor growth control achieved in a significant proportion of VIPoma patients (42, 55, 56)(15, 16, 43).

REFERENCES

 

  1. Larsson LI, Fahrenkrug J, Schaffalitzky De Muckadell O, Sundler F, Håkanson R, Rehfeld JR. Localization of vasoactive intestinal polypeptide (VIP) to central and peripheral neurons. Proc Natl Acad Sci U S A. 1976;73(9):3197-200.
  2. Holst JJ, Fahrenkrug J, Knuhtsen S, Jensen SL, Poulsen SS, Nielsen OV. Vasoactive intestinal polypeptide (VIP) in the pig pancreas: role of VIPergic nerves in control of fluid and bicarbonate secretion. Regul Pept. 1984;8(3):245-59.
  3. Robberecht P, Conlon TP, Gardner JD. Interaction of porcine vasoactive intestinal peptide with dispersed pancreatic acinar cells from the guinea pig. Structural requirements for effects of vasoactive intestinal peptide and secretin on cellular adenosine 3':5'-monophosphate. J Biol Chem. 1976;251(15):4635-9.
  4. Barbezat GO, Grossman MI. Intestinal secretion: stimulation by peptides. Science. 1971;174(4007):422-4.
  5. Krejs GJ. VIPoma syndrome. Am J Med. 1987;82(5b):37-48.
  6. Verner JV, Morrison AB. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med. 1958;25(3):374-80.
  7. Pieterman CRC, van Leeuwaarde RS, van den Broek MFM, van Nesselrooij BPM, Valk GD. Multiple Endocrine Neoplasia Type 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  8. Said SI, Mutt V. Potent peripheral and splanchnic vasodilator peptide from normal gut. Nature. 1970;225(5235):863-4.
  9. Mutt V, Said SI. Structure of the porcine vasoactive intestinal octacosapeptide. The amino-acid sequence. Use of kallikrein in its determination. Eur J Biochem. 1974;42(2):581-9.
  10. Said SI, Mutt V. A peptide fraction from lung tissue with prolonged peripheral vasodilator activity. Scand J Clin Lab Invest Suppl. 1969;107:51-6.
  11. Bloom SR, Polak JM, Pearse AG. Vasoactive intestinal peptide and watery-diarrhoea syndrome. Lancet. 1973;2(7819):14-6.
  12. Kane MG, O'Dorisio TM, Krejs GJ. Production of secretory diarrhea by intravenous infusion of vasoactive intestinal polypeptide. N Engl J Med. 1983;309(24):1482-5.
  13. Kapoor R, Moseley RH, Kapoor JR, Crapo LM, Saint S. Clinical problem-solving. Needle in a haystack. N Engl J Med. 2009;360(6):616-21.
  14. Schizas D, Mastoraki A, Bagias G, Patras R, Moris D, Lazaridis, II, et al. Clinicopathological data and treatment modalities for pancreatic vipomas: a systematic review. J buon. 2019;24(2):415-23.
  15. Angelousi A, Koffas A, Grozinsky-Glasberg S, Gertner J, Kassi E, Alexandraki K, et al. Diagnostic and Management Challenges in Vasoactive Intestinal Peptide Secreting Tumors: A Series of 15 Patients. Pancreas. 2019;48(7):934-42.
  16. Brugel M, Walter T, Goichot B, Smith D, Lepage C, Do Cao C, et al. Efficacy of treatments for VIPoma: A GTE multicentric series. Pancreatology. 2021;21(8):1531-9.
  17. Mete O, Basturk O, Brosens LA, de Herder W, Hong SM, La Rosa S. VIPoma. In: Klimstra DS, Osamura RY, editors. WHO Classification of Tumours Endocrine and neuroendocrine tumours. Lyon: International Agency for Research on Cancer; 2023.
  18. Siddappa PK, Vege SS. Vasoactive Intestinal Peptide-Secreting Tumors: A Review. Pancreas. 2019;48(9):1119-25.
  19. Soga J, Yakuwa Y. Vipoma/diarrheogenic syndrome: a statistical evaluation of 241 reported cases. J Exp Clin Cancer Res. 1998;17(4):389-400.
  20. Kamp K, Feelders RA, van Adrichem RC, de Rijke YB, van Nederveen FH, Kwekkeboom DJ, et al. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab. 2014;99(9):3060-9.
  21. Kaltsas G, Androulakis, II, de Herder WW, Grossman AB. Paraneoplastic syndromes secondary to neuroendocrine tumours. Endocr Relat Cancer. 2010;17(3):R173-93.
  22. Halfdanarson TR, Strosberg JR, Tang L, Bellizzi AM, Bergsland EK, OʼDorisio TM, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(7):863-81.
  23. Lévy-Bohbot N, Merle C, Goudet P, Delemer B, Calender A, Jolly D, et al. Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterol Clin Biol. 2004;28(11):1075-81.
  24. Niederle B, Selberherr A, Bartsch D, Brandi ML, Doherty GM, Falconi M, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) and the Pancreas - Diagnosis and Treatment of Functioning and Non-Functioning Pancreatic and Duodenal Neuroendocrine Neoplasia within the MEN1 Syndrome - An International Consensus Statement. Neuroendocrinology. 2020.
  25. Smith SL, Branton SA, Avino AJ, Martin JK, Klingler PJ, Thompson GB, et al. Vasoactive intestinal polypeptide secreting islet cell tumors: a 15-year experience and review of the literature. Surgery. 1998;124(6):1050-5.
  26. Ghaferi AA, Chojnacki KA, Long WD, Cameron JL, Yeo CJ. Pancreatic VIPomas: subject review and one institutional experience. J Gastrointest Surg. 2008;12(2):382-93.
  27. Leibowitz-Amit R, Mete O, Asa SL, Ezzat S, Joshua AM. Malignant pheochromocytoma secreting vasoactive intestinal peptide and response to sunitinib: a case report and literature review. Endocr Pract. 2014;20(8):e145-50.
  28. Murahashi N, Abe T, Matsumoto R, Oosawa T, Yoshinaga K, Shiga T, et al. [A Case of WDHA Water Diarrhea Hypokalemia Achlorhydria Syndrome that Developed after Multimodal Therapy for Retroperitoneal Paraganglioma]. Hinyokika Kiyo. 2019;65(7):277-82.
  29. Onozawa M, Fukuhara T, Minoguchi M, Takahata M, Yamamoto Y, Miyake T, et al. Hypokalemic rhabdomyolysis due to WDHA syndrome caused by VIP-producing composite pheochromocytoma: a case in neurofibromatosis type 1. Jpn J Clin Oncol. 2005;35(9):559-63.
  30. Ikuta S, Yasui C, Kawanaka M, Aihara T, Yoshie H, Yanagi H, et al. Watery diarrhea, hypokalemia and achlorhydria syndrome due to an adrenal pheochromocytoma. World J Gastroenterol. 2007;13(34):4649-52.
  31. Hanna FW, Ardill JE, Johnston CF, Cunningham RT, Curry WJ, Russell CF, et al. Regulatory peptides and other neuroendocrine markers in medullary carcinoma of the thyroid. J Endocrinol. 1997;152(2):275-81.
  32. Said SI. Evidence for secretion of vasoactive intestinal peptide by tumours of pancreas, adrenal medulla, thyroid and lung: support for the unifying APUD concept. Clin Endocrinol (Oxf). 1976;5 Suppl:201s-4s.
  33. Said SI, Faloona GR. Elevated plasma and tissue levels of vasoactive intestinal polypeptide in the watery-diarrhea syndrome due to pancreatic, bronchogenic and other tumors. N Engl J Med. 1975;293(4):155-60.
  34. Bloom SR. Vasoactive intestinal peptide, the major mediator of the WDHA (pancreatic cholera) syndrome: value of measurement in diagnosis and treatment. Am J Dig Dis. 1978;23(4):373-6.
  35. Domschke S, Domschke W, Bloom SR, Mitznegg P, Mitchell SJ, Lux G, et al. Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects. Gut. 1978;19(11):1049-53.
  36. Modlin IM, Bloom SR, Mitchell S. Plasma vasoactive intestinal polypeptide (VIP) levels and intestinal ischaemia. Experientia. 1978;34(4):535-6.
  37. Doherty CC, Buchanan KD, Ardill J, McGeown MG. Elevations of gastrointestinal hormones in chronic renal failure. Proc Eur Dial Transplant Assoc. 1978;15:456-65.
  38. Clark AJ, Adrian TE, McMichael HB, Bloom SR. Vasoactive intestinal peptide in shock and heart failure. Lancet. 1983;1(8323):539.
  39. Nikou GC, Toubanakis C, Nikolaou P, Giannatou E, Safioleas M, Mallas E, et al. VIPomas: an update in diagnosis and management in a series of 11 patients. Hepatogastroenterology. 2005;52(64):1259-65.
  40. de Herder WW, Zandee WT, Hofland J. Insulinoma. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  41. de Mestier L, Hentic O, Cros J, Walter T, Roquin G, Brixi H, et al. Metachronous hormonal syndromes in patients with pancreatic neuroendocrine tumors: a case-series study. Ann Intern Med. 2015;162(10):682-9.
  42. Crona J, Norlén O, Antonodimitrakis P, Welin S, Stålberg P, Eriksson B. Multiple and Secondary Hormone Secretion in Patients With Metastatic Pancreatic Neuroendocrine Tumours. J Clin Endocrinol Metab. 2016;101(2):445-52.
  43. Marques B, Monteiro AR, Martins RG, Couto J, Rodrigues F, Ribeiro J. Metastatic VIPoma, Cosecreting Insulin, With Complete Response to Lanreotide, Capecitabine, and Temozolomide. Pancreas. 2020;49(3):e19-e20.
  44. Ito T, Jensen RT. Perspectives on the current pharmacotherapeutic strategies for management of functional neuroendocrine tumor syndromes. Expert Opin Pharmacother. 2021;22(6):685-93.
  45. O'Dorisio TM, Mekhjian HS, Gaginella TS. Medical therapy of VIPomas. Endocrinol Metab Clin North Am. 1989;18(2):545-56.
  46. Kaltsas G, Caplin M, Davies P, Ferone D, Garcia-Carbonero R, Grozinsky-Glasberg S, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pre- and Perioperative Therapy in Patients with Neuroendocrine Tumors. Neuroendocrinology. 2017;105(3):245-54.
  47. Frilling A, Modlin IM, Kidd M, Russell C, Breitenstein S, Salem R, et al. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15(1):e8-21.
  48. Kunz PL, Reidy-Lagunes D, Anthony LB, Bertino EM, Brendtro K, Chan JA, et al. Consensus guidelines for the management and treatment of neuroendocrine tumors. Pancreas. 2013;42(4):557-77.
  49. Pavel M, O'Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016;103(2):172-85.
  50. Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(7):844-60.
  51. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224-33.
  52. Dromain C, Loaiza-Bonilla A, Mirakhur B, Beveridge TJR, Fojo AT. Novel Tumor Growth Rate Analysis in the Randomized CLARINET Study Establishes the Efficacy of Lanreotide Depot/Autogel 120 mg with Prolonged Administration in Indolent Neuroendocrine Tumors. Oncologist. 2021;26(4):e632-e8.
  53. O'Dorisio TM, Anthony LB. A 25-Year Experience of Gastroenteropancreatic Neuroendocrine Tumors and Somatostatin (Congeners) Analogs: From Symptom Control to Antineoplastic Therapy. Front Horm Res. 2015;44:177-92.
  54. Chen C, Zheng Z, Li B, Zhou L, Pang J, Wu W, et al. Pancreatic VIPomas from China: Case reports and literature review. Pancreatology. 2019;19(1):44-9.
  55. Yao JC, Pavel M, Lombard-Bohas C, Van Cutsem E, Voi M, Brandt U, et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers From the Randomized, Phase III RADIANT-3 Study. J Clin Oncol. 2016;34(32):3906-13.
  56. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514-23.
  57. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501-13.
  58. Fazio N, Kulke M, Rosbrook B, Fernandez K, Raymond E. Updated Efficacy and Safety Outcomes for Patients with Well-Differentiated Pancreatic Neuroendocrine Tumors Treated with Sunitinib. Target Oncol. 2021;16(1):27-35.
  59. Bourcier ME, Vinik AI. Sunitinib for the treatment of metastatic paraganglioma and vasoactive intestinal polypeptide-producing tumor (VIPoma). Pancreas. 2013;42(2):348-52.
  60. de Mestier L, Walter T, Brixi H, Lombard-Bohas C, Cadiot G. Sunitinib achieved fast and sustained control of VIPoma symptoms. Eur J Endocrinol. 2015;172(1):K1-3.
  61. Anderson JV, Bloom SR. Treatment of malignant endocrine pancreatic tumours with human leucocyte interferon. Lancet. 1987;1(8524):97.
  62. Oberg K, Alm G, Lindström H, Lundqvist G. Successful treatment of therapy-resistant pancreatic cholera with human leucocyte interferon. Lancet. 1985;1(8431):725-7.
  63. Barraclough MA, Bloom SR. Vipoma of the pancreas: observations on the diarhrhea and circulatory disturbances. Arch Intern Med. 1979;139(4):467-71.
  64. Brabander T, van der Zwan WA, Teunissen JJM, Kam BLR, Feelders RA, de Herder WW, et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin Cancer Res. 2017;23(16):4617-24.
  65. Zandee WT, Brabander T, Blazevic A, Kam BLR, Teunissen JJM, Feelders RA, et al. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104(4):1336-44.
  66. Audil HY, Eiring RA, Kendi AT, Halfdanarson TR. A Case of Metastatic VIPoma With Complete Response to Peptide Radionuclide Receptor Therapy. Pancreas. 2021;50(4):e45-e6.
  67. Dréanic J, Lepère C, El Hajjam M, Gouya H, Rougier P, Coriat R. Emergency therapy for liver metastases from advanced VIPoma: surgery or transarterial chemoembolization? Ther Adv Med Oncol. 2016;8(5):383-7.
  68. Hengst K, Nashan B, Avenhaus W, Ullerich H, Schlitt HJ, Flemming P, et al. Metastatic pancreatic VIPoma: deteriorating clinical course and successful treatment by liver transplantation. Z Gastroenterol. 1998;36(3):239-45.
  69. Bramley PN, Lodge JP, Losowsky MS, Giles GR. Treatment of metastatic Vipoma by liver transplantation. Clin Transplant. 1990;4(5 part 1):276-8; discussion 9.
  70. Johnston PC, Ardill JE, Johnston BT, Mc Cance DR. Vasoactive intestinal polypeptide secreting pancreatic tumour with hepatic metastases: long term survival after orthotopic liver transplantation. Ir J Med Sci. 2010;179(3):439-41.

 

Insulinoma

ABSTRACT

 

Insulinomas are rare pancreatic neuroendocrine neoplasms (panNENs - incidence of 1-3 cases per million per year). Most are solitary and do not show signs of malignant spread. Multiple synchronous or metachronous panNENs / insulinomas may occur in multiple endocrine neoplasia type 1 (MEN-1). The diagnosis of an insulinoma requires demonstration of inappropriately high insulin, proinsulin or C-peptide levels for the prevailing hypoglycemia in a 72h fast. Localization of the tumor and exclusion or confirmation of metastatic disease by computed tomography is still the preferred initial option followed by endoscopic ultrasonography (EUS) or MRI. Glucagon-like peptide receptor 1 (GLP-1R) receptor positron emission tomography (PET) CT or MRI is a highly sensitive localization technique for indolent, localized (“benign”) insulinomas. For single solitary tumors surgical excision or radiofrequency ablation are the treatments of choice. In aggressive malignant (metastatic) cases, debulking of the panNENs, including locoregional lymph nodes can be considered. If hyperinsulinemia and hypoglycemia persist, diazoxide with a thiazide diuretic relieves hypoglycemia. Liver metastases can be resected, or treated by bland embolization, radioembolization (SIRT), radiofrequency ablation (RFA), microwave and cryoablation, high-intensity focused ultrasound (HIFU), laser, brachytherapy and irreversible electroporation (IRE) depending on local availability. In patients with unresectable low-grade metastatic malignant insulinomas, the long-acting somatostatin analog Lanreotide Autogel is the approved first-line therapy for control of tumor growth and sometimes control of hypoglycemia is achieved with this drug. If indicated, peptide receptor radiotherapy (PRRT) with radiolabeled somatostatin analogs, or Everolimus can be used for tumor, symptom and glucose control. Malignant NENs can also be treated with cytotoxic chemotherapy regimens, particularly those with a high tumor grade.

 

HISTORY

 

The pancreatic islet cells were first described by the German medical student Paul Langerhans (1847-1888) in 1869. James R Macleod (1876-1935), Frederick G. Banting (1891-1941), Charles H. Best (1899-1978) and James B. Collip (1892-1965) first isolated insulin in 1922. The US surgeon Seale Harris (1870-1957) was the first to identify a case of endogenous hyperinsulinism. In 1926, the US surgeon William J Mayo (1861-1939) performed an exploratory laparotomy on a patient with recurrent severe hypoglycemia and found an unresectable pancreatic tumor (malignant insulinoma) with multiple liver, lymph node, and mesenteric metastases. In 1927, the US physician Russel M. Wilder (1885-1959) and colleagues reported on the necropsy of this patient. Extracts of a liver metastasis produced marked lowering of the blood glucose levels when injected into rabbits. It seems also likely that Mayo’s & Wilder’s patient had multiple endocrine neoplasia type 1 (MEN-1) since he also had renal stones and his cousin had had similar symptoms. In 1954, the US internist Paul Wermer (1898-1975) reported disorders of one or more endocrine glands in five members of one family in 1954. This familial syndrome was once called Wermer syndrome, but is nowadays better known as multiple endocrine neoplasia type 1 (MEN-1).

 

The first cure of hyperinsulinism by removal of an insulinoma by the Canadian surgeon Roscoe R. Graham (1890-1948) was reported in 1929 by Goldwin Howland (1875-1950) and co-workers. The US surgeon Allen O. Whipple (1881-1963) and pathologist Virginia K. Frantz (1896-1967) identified the diagnostic hallmark of insulinoma better known as “Whipple’s triad” (1).

 

INTRODUCTION

 

More than 99% of insulinomas are located in the pancreas (2, 3). Extremely rare extra-pancreatic (metastatic) insulinomas have been described in the lung, duodenum, ileum, jejunum, hilum of the spleen, and gastric antrum (4-9). Insulinomas are the most common hormone-producing neuroendocrine neoplasms (NENs) of the pancreas, with an estimated incidence of 1–3 per million per year. Insulinomas are evenly distributed in the pancreas (2, 3). There is an age-specific incidence peak in the fifth decade of life and the incidence is slightly higher in women than in men. Approximately 10% are multiple, 10-15% show malignant spread. As the definitions for malignancy are ambiguous, non-metastatic insulinomas are nowadays referred to as "indolent" and metastatic insulinomas as "aggressive" (3, 10). Patients with aggressive insulinoma have lower survival compared to patients with indolent insulinoma: 5-year-survival has been reported to be 94.5-100% for indolent and 24-66.8% for aggressive disease (3, 11-13).

 

After initial recognition of the key symptoms, careful laboratory testing, sophisticated imaging and eventually meticulous surgery follows in most cases. It is evident that a multidisciplinary team (MDT) approach is required. The hallmark features of insulinomas resulting from hypoglycemia include neuroglycopenic (e.g., confusion, visual changes, unusual behavior) and sympathetico-adrenal (e.g., palpitations, diaphoresis, tremulousness) symptoms. A firmly established diagnosis of an insulin-secreting lesion of the pancreas is essential for successful management. Therefore, it is critically important to rule out other causes of hypoglycemia associated with fasting (13, 14).

 

HEREDITARY TUMORS

 

An overview of the multiple endocrine neoplasia type 1 (MEN1) syndrome can be found in the chapter “MEN1 (15). Fifty percent of MEN-1 patients harbor pancreatic NENs (panNENs) (13, 15, 16). 5–10% of insulinomas are associated with the MEN1 syndrome. MEN1-related NENs / insulinomas may occur as multiple lesions (15). In patients with the multiple endocrine neoplasia type 4 (MEN4) syndrome caused by inactivating mutations in the CDKN1B (Cyclin Dependent Kinase Inhibitor 1B) gene, pancreatic NENs can also be found, but it is unclear if insulinomas are more prevalent in MEN4 (17, 18). PanNENs can also be diagnosed in patients with von Hippel Lindau disease (VHL), but also there seems not to be a preponderance of insulinomas in this syndrome (19). Tuberous sclerosis complex (TSC) is a genetic tumor-predisposing syndrome associated with the development of multiple hamartomas among other abnormalities. TSC is caused by mutations of two tumor suppressor genes, TSC1 on chromosome 9q34 and TSC2 on chromosome 16p13.3, which encode for hamartin and tuberin, respectively. PanNENs are uncommon in TSC, but insulinoma seems to be the predominant panNEN in this genetic disorder (20).

 

CLINICAL FEATURES

 

The hallmark of the diagnosis of insulinoma is Whipple’s triad: 1) symptoms known or likely to be caused by hypoglycemia, 2) a low plasma glucose measured at the time of the symptoms and 3) relief of symptoms when the glucose is raised back to normal. The principal biochemical feature of an insulinoma is hypoglycemia but there are other malignancies and disorders which can cause hypoglycemia like big-IGF-2-producing tumors, glycogen storage diseases, administration of exogenous insulin or oral glucose-lowering drugs, insulinomatosis, the autoimmune insulin antibody syndrome (Hirata’s disease) or insulin receptor (anti-ISR) antibody syndrome (Flier’s syndrome) and congenital hyperinsulinism/nesidioblastosis in the pancreas (14, 21-31). While hypoglycemia is a hallmark of insulinoma, the low blood glucose level alone is not diagnostic of insulinoma, nor in general is the absolute insulin level elevated in all cases of organic hyperinsulinism. Hypoglycemia activates the adrenergic and cholinergic nervous systems and depending on the degree of the hypoglycemia presents different levels of impairment of neurologic function (Table 1) (14, 29, 32-35).

 

Table 1. Distinguishing Signs and Symptoms of Insulinomas

Neurogenic

Neuroglycopenic

·    Adrenergic

Palpitations

Tremor

Anxiety/arousal/nervousness

·    Cholinergic

Sweating/diaphoresis

Hunger

Paresthesia

Circumpolar tingling

· Blurred Vision

· Cognitive impairments

· Behavioral changes

· Psychomotor abnormalities

· Confusion

· Disorientation

· Memory Loss

· Seizure

· Stupor

 

BIOCHEMICAL DIAGNOSIS

 

The first step in the diagnosis of an insulinoma is to demonstrate hyperinsulinemic hypoglycemia (this is also called “organic hyperinsulinism”). This can potentially be achieved during a spontaneous hypoglycemia. However, most frequently a 72-hour fast is needed, which is currently the standard test to diagnose an insulinoma. The patient is closely clinically observed while serial glucose and insulin levels are obtained over the 72 hours until the patient becomes symptomatic, or a hypoglycemia is demonstrated. More than 95% of cases can be diagnosed based on responses to this easy test. Because the absolute insulin level is not elevated in all patients with insulinomas, a nondetectable or nonelevated insulin level does not rule out the disease. Values of insulin equal to or greater than 3 μU/mL (using modern insulin assays) in the presence of a blood glucose less than 3 mmol/l (55 mg/dl) are highly suggestive. Most specialists prefer more stringent cut-off glucose values amounting to 2.2 – 2.5 mmol/L (40 - 45 mg/dL) or less to increase the diagnostic specificity. Because of the potential increased proinsulin secretion, which is not detected using the currently used insulin assays, it is generally recommended also to measure proinsulin and/or C-peptide levels, particularly in those cases with low to undetectable insulin levels in the blood. In the past these elevated proinsulin levels were also detected using the insulin RIAs, whereas nowadays these tumors are inadvertently addressed as pro-insulinomas. In these cases, concomitant C-peptide levels equal to or greater than 0.2 nmol/l and/or concomitant pro-insulin levels equal to or greater than 5 pmol/l (in the presence of a hypoglycemia) are also suggestive of an insulinoma. Commercial insulin preparations do not contain C-peptide and low C-peptide levels combined with high insulin levels confirm the diagnosis of factitious hyperinsulinemia (14, 21, 29, 36, 37).

 

Furthermore, absence of sulfonylurea (metabolites) in the plasma or urine has also been used to exclude factitious hypoglycemia’s in (von) Munchhausen syndrome / (von) Munchausen by proxy. Patients who take sulfonylureas surreptitiously may have raised insulin and C-peptide values soon after ingestion, but chronic use will result in hypoglycemia without raised insulin or C-peptide levels. Only a high index of suspicion and measurement of plasma or urine sulfonylureas will lead to the correct diagnosis. (14, 21, 29, 37).

 

Finally, the demonstration of ß-hydroxy-butyrate levels equal to or less than 2.7 mmol/l at end of fast is used by some to confirm the hyperinsulinemic state. Some experts require the demonstration of a glucose response to 1 mg glucagon of more than 1.4 mmol/l (25 mg/dl) at end of fast. This increase of glucose is illustrative for the hyperinsulinemic state, because hyperinsulinemia preserves the liver glycogen storage despite (14, 21, 24, 29, 32-37).

 

TUMOR LOCALIZATION

 

Once the diagnosis of insulinoma is confirmed, every effort should be made to localize the tumor. Preoperative localization is important because approximately 30% of insulinomas are less than 1 cm in diameter and 10% are multiple, the latter particularly is present in MEN-1 patients (16). In addition, 10 to 15% are aggressive, malignant (metastatic), and very few patients will have either islet cell hyperplasia, or congenital hyperinsulinism/nesidioblastosis and no visible tumor at all. The anatomical localization of nonmetastatic (benign) insulinomas is also important for the choice between laparoscopic, robot-assisted, and open pancreatic surgery and between enucleation or resection – partial pancreatectomy and radiofrequency ablation (RFA) (37). Techniques most commonly used to demonstrate tumors in the pancreas include 3 phase CT and MRI, and endoscopic ultrasound (EUS). Each modality has variable reported abilities to identify insulinomas, likely reflecting institutional or operator-dependent (like in EUS) expertise (Table 2) (37).

 

Table 2. Imaging Strategies in Insulinoma Patients

 

Sensitivity

Transabdominal ultrasound                           

Three phase CT                                               

MRI (T1 +T2 weighted images + fat suppression)

Endoscopic Ultrasound (EUS)                                  

Arterial Calcium Stimulation - Venous Sampling      

9 -65%

60-80%

85-90%

75-90%

80-90%

Intraoperative Localizing Techniques

Palpation                                                        

Intraoperative ultrasound (IOUS)                  

Palpation plus IOUS  

 

70%

75-90%

85-95%

Nuclear Medicine

Somatostatin receptor scintigraphy SPECT / PET*

18F-DOPA PET                                                           

Glucagon-Like Peptide-1 (Exendin-4) Receptor Imaging SPECT / PET**

 

46-50% / 50-86%

50%

75 / 95%

*, preferably used in patients with aggressive – malignant – metastatic insulinomas

**, preferably used in patients with indolent – (“benign”) – localized insulinomas

 

In the past, selective pancreatic angiography and elective intra-arterial injection of calcium with sampling of hepatic vein insulin were used on a regular basis in high volume centers (38, 39). These invasive regionalization (an exact localization will be never given) procedures became less used because of the improved imaging procedures mentioned above and the introduction of glucagon-like peptide 1 (GLP-1) receptor imaging. The glucagon-like peptide 1 receptor (GLP-1R) is mainly expressed on the pancreatic beta cells and is therefore an interesting target for imaging of (previously occult) indolent (“benign”) localized insulinomas. However, as opposed to localized, indolent (“benign”) insulinomas, aggressive malignant (metastatic) insulinomas often lack the GLP-1R. Conversely, malignant (metastatic) aggressive insulinomas often do express the somatostatin receptor subtype 2 (SST2), which can be targeted using PET/CT or PET/MRI using 68Ga-DOTA-labeled somatostatin analogs (SSAs) or in the past with somatostatin receptor scintigraphy and SPECT (40) (11, 41). In various studies, the GLP-1 receptor agonists 111In-DOTA-exendin-4 and/or 68Ga-DOTA-exendin-4 PET/CT successfully detected localized indolent (“benign”) insulinomas. 68Ga-DOTA-exendin-4 PET/CT seems more sensitive than 111In-DOTA-exendin-4 SPECT/CT (41, 42). Replacing DOTA by NODAGA for 68Ga-NODAGA-exendin-4 PET/CT ensures higher specific activities (Figure 1).

 

Figure 1. Localization studies demonstrating a localized insulinoma. From left to right: arterial-phase contrast-enhanced CT, 68Ga-DOTATATE PET-CT, 68Ga-NODAGA-exendin PET-CT (Courtesy: Drs. Marti Boss and Martin Gotthardt, Radboud University Medical Centre, Nijmegen, the Netherlands).

The efficacy of fluorine-18-L-3,4-dihydroxyphenylalanine (18F-DOPA) PET/CT is based on co-secretion of dopamine and hormones or peptides by NEN cells. In these cells, L-DOPA is converted by the enzyme L-DOPA decarboxylase to dopamine. Next to  68Ga-NODAGA-exendin-4 PET/CT (43), 18F-DOPA PET/CT (with carbidopa premedication) plays an important role in the differential diagnosis of congenital hyperinsulinism (nesidioblastosis), especially for the identification of focal forms (28, 43-45).

 

If all localization and regionalization techniques fail to localize a tumor, intraoperative palpation of the pancreas and intraoperative ultrasound might prove to be successful (46).

 

In addition to the assessment of insulin hypersecretion, the metastatic spread, as reflected by the (ENETS/AJCC-UICC) staging, also determines the clinical manifestations and contribute to the prognosis (Figure 2 and Table 3) (28-31). Secondary, or metachronous insulin secretion by pancreatic neuroendocrine tumors which previously were non-secreting, or secreted other peptide hormones can also occur and is generally associated with poor survival (47, 48).

Figure 2. TNM staging system for pancreatic neuroendocrine tumors including insulinomas.

 

Table 3. TNM Staging System for Pancreatic Neuroendocrine Tumors including Insulinomas

Stage

T

N

M

I

T1

N0

M0

IIa

T2

N0

M0

IIb

T3

N0

M0

IIIa

T4

N0

M0

IIIb

Any T

N1

M0

IV

Any T

Any N

M1

 

HISTOPATHOLOGY

 

The WHO classification and grading of panNENs separates these tumors using the Ki67 index (MIB-1 antibody staining) into 4 broad categories: grade 1-2 (G1-2) well-differentiated pancreatic NETs (panNETs), poorly differentiated pancreatic neuroendocrine carcinomas (NECs – panNECs) and well-differentiated grade 3 (G3) NET. Helpful for the distinction of NECs from G3 NETs is their overexpression of p53 and loss of expression of Rb1 (Table 4). Insulin staining is not obligatory positive in insulinomas and is usually not necessarily required once the clinical diagnosis is made (3, 10, 49, 50).

 

Table 4. WHO 2017/2023 Classification for Neuroendocrine Neoplasms (NENs) of the Pancreas

Differentiation

Name       Grade

Ki 67 (% of ≥500 cells)

Mitotic count (2 mm2)

Well differentiated

NET            G1

                   G2

                   G3

<3

3-20

>20

<2

2-20

>20

Poorly differentiated

NEC          (G3)

Small cell type

Large cell type

>20

>20

 

Indolent and aggressive insulinoma are different entities. Aggressive insulinomas are characterized by rapid onset of symptoms, larger size, expression of ARX and alpha-1-antitrypsin; and decreased or absent immunohistochemical expression of insulin, PDX1 and GLP-1R. Moreover, aggressive insulinomas often harbor Alpha-Thalassemia/mental Retardation, X-linked (ATRX) and Death Domain Associated Protein (DAXX) mutations, the alternative lengthening of telomeres phenotype (ALT) and chromosomal instability (CIN). Tumor grade and MEN1 and YY1 mutations are less useful for predicting behavior. Aggressive insulinomas have similarities to normal alpha-cells and nonfunctional pancreatic neuroendocrine tumors, while indolent insulinomas remain closely related to normal beta-cells (11, 51),

 

SURGICAL AND INTERVENTIONAL TREATMENT

 

The treatment of pancreatic localized insulinoma usually is surgical; in the great majority of cases, it will provide a complete cure. It should be performed only when the diagnosis is certain, however, and by a surgeon who is skilled in pancreatic surgery. The surgical approach to an insulinoma is straightforward when the tumor is localized. Localized insulinomas are typically removed by enucleation of the tumor and rarely do tumors at the head of the pancreas require a pancreaticoduodenectomy (Whipple’s procedure). Precise localization obviates blind pancreatic resection. EUS with special focus on the relationship between the tumor and the pancreatic duct is an excellent tool to guide the surgical decision. Laparoscopic, or robot-assisted enucleation of an insulinoma has been shown to be feasible, particularly if the lesion is visualized pre-operatively on CT scan or by EUS. In patients who have been unresponsive to medical therapy and in whom 18F-DOPA PET/CT, PTHVS, or intra-arterial calcium stimulation with venous sampling suggests diffuse or multiple sources, such as adenomatosis, nesidioblastosis/congenital hyperinsulinemia, or hyperplasia, a resection of at least 80% of the distal pancreas can be indicated. In selected cases curative endoscopic ultrasound-guided radiofrequency ablation (EUS-RFA) of a localized insulinoma can be feasible (2, 46, 52-54).

 

Malignant aggressive (metastatic) insulinomas can occasionally be surgically cured when there is localized or oligometastatic disease. Also, liver metastases can be resected, or treated by bland or chemo-embolization (TACE), radioembolization (SIRT), radiofrequency ablation (RFA), microwave and cryoablation, high-intensity focused ultrasound (HIFU), laser, brachytherapy and irreversible electroporation (IRE) depending on availability at the institution (55). If more than 90% of tumor load can be resected, palliative surgery can also be considered. However, most aggressive malignant metastatic insulinomas cannot be cured by surgery only and require medical antihormonal and antitumor treatment (46).

 

MEDICAL MANAGEMENT

 

When hypoglycemia can be controlled with diet alone or with small, well-tolerated doses of diazoxide, and/or when the medical condition of the patient increases the hazard of surgery sufficiently, medical management alone may be considered. Patients with diffuse hyperinsulinism for whom an operation is planned first should have a trial of treatment with diazoxide and a natriuretic benzothiadiazide. Medical treatment is required for the great majority of malignant insulinomas because only occasionally are they cured by operation. Medical treatment for localized, indolent (“benign”) insulinomas includes a change in meals to include “lente carbohydrate” or unrefined carbohydrate given as frequently as required to prevent hypoglycemia. The management of malignant insulinoma is antihormonal and antitumor therapy (14, 46). 

 

DIETARY MANAGEMENT

 

The cornerstone of medical management of insulinoma and other forms of hyperinsulinism is the diet. Not uncommonly, patients may avoid symptoms of hypoglycemia for variable periods of time by shortening the number of hours between meals. For some, the inclusion of a bedtime (11:00 pm) feeding is sufficient; for others, a midmorning, midafternoon, and/or a 3:00 pm snack is necessary. More slowly absorbable forms of carbohydrates (e.g., starches, bread, potatoes, rice) generally are preferred. During hypoglycemic episodes, rapidly absorbable forms (e.g., fruit juices with added glucose or sucrose) are indicated. In patients with severe refractory hypoglycemia, use of a continuous nasogastric tube feeding or intravenous infusion of glucose, coupled with increased dietary intake of carbohydrate, frequently alleviates hypoglycemia long enough to institute additional therapy (14).

 

MEDICAL THERAPY

 

Diazoxide (Proglycem) owes its potent hyperglycemic properties to two effects: it directly inhibits the release of insulin by β cells through stimulation of α-adrenergic receptors. It also has an extra-pancreatic hyperglycemic effect, probably by inhibiting cyclic adenosine monophosphate phosphodiesterase (cyclic AMP), resulting in higher plasma levels of cyclic AMP and enhanced glycogenolysis. Because diazoxide induces the retention of sodium, edema is troublesome at higher dosages. The addition of a diuretic benzothiadiazine (e.g., hydrochlorothiazide) not only corrects or prevents edema but synergizes the hyperglycemic effect of diazoxide. At the doses needed to counteract the higher doses of diazoxide (e.g., 450-600 mg/d), natriuretic benzothiadiazines frequently induce hypokalemia. Nausea is an additional complication at higher dosages of diazoxide, and hypertrichosis may complicate long-term treatment. These compounds have been useful to elevate blood levels of glucose into the euglycemic range if an operation must be delayed for weeks or months. If they can be tolerated, higher doses may be used in patients with malignant insulinomas (56).

 

Theoretically, calcium channel blockers are capable of inhibiting insulin secretion. Verapamil and diltiazem have been used with variable results in patients with organic hyperinsulinism (57, 58).

 

β-Adrenergic-receptor blocking drugs inhibit insulin secretion and therefore may be of value in treating organic hyperinsulinism. The use of propranolol has been associated with the reduction of plasma insulin levels and with the relief of hypoglycemic attacks in patients with localized, indolent (“benign”), or aggressive malignant (metastatic) insulinoma. Because this drug can also mask the adrenergic symptoms of hypoglycemia and inhibit muscle glycogenolysis, however, there is a risk of aggravating the clinical syndrome. The drug should be used with extreme caution and careful monitoring (59).

 

The anticonvulsive diphenylhydantoin has been shown to inhibit the in vitro release of insulin from both the labile and storage β-cell pools. In only one-third or less of patients with localized, indolent (“benign”) insulinoma, however, is the hyperglycemic effect of diphenylhydantoin of any clinical significance (60, 61). Furthermore, adverse effects usually occur with the dosages required. Maintenance doses range from 300 to 600 mg/d. The concurrent administration of diazoxide lowers measurable blood levels of diphenylhydantoin, and their concurrent use is not recommended.

 

Several reports exist on the successful use of intermediate acting subcutaneous octreotide injections (100-500 µg t.i.d.) in prolonging the ability to fast in a patient with localized, indolent (“benign”) and aggressive malignant (metastatic) insulinomas. However, long-term administration of depot octreotide (Sandostatin LAR 30 mg / 4wks IM) or lanreotide (Somatuline Autogel 120 mg / 4 wks deep SC) may give only short-term relief of hypoglycemia. SSAs may also actually worsen plasma glucose levels probably by inhibiting the counterregulatory glucagon response. SSA treatment in insulinoma and nesidioblastosis patients should, therefore, always be preceded by a clinical trial with intermediate acting subcutaneous octreotide. In a limited number of cases, the second generation pan-SSA pasireotide has been successfully used to control hypoglycemias in patients with malignant insulinomas (62-65).

 

Targeting the pathway of the mammalian target of rapamycin (mTOR) has been shown in several trials to be effective in the management of low grade metastatic inoperable neuroendocrine tumors (66). Several studies have recently shown that everolimus (10mg/day) can normalize blood glucose levels in insulinoma patients. mTOR inhibitors like everolimus can reduce the insulin secretion and increase insulin resistance (62, 67-72). The multi-kinase inhibitor sunitinib (25mg/day) has only been occasionally reported to improve symptoms of hypoglycemia (62, 68, 73). Tyrosine kinase inhibitors (TKIs) do not have the capacity to suppress insulin, as well as inducing insulin resistance, like everolimus.

 

The use of glucocorticoids, which increase gluconeogenesis and cause insulin resistance, also can help to stabilize blood glucose at an acceptable level. Pharmacologic doses (Prednisone, approximately 1 mg/kg) must be used (74). Glucagon may help to raise blood glucose concentrations, but it may simultaneously directly stimulate the release of insulin (55).

 

ANTI-TUMOR TREATMENT IN MALIGNANT INSULINOMAS

 

Like in the other panNEN subtypes, anti-tumor treatments can consist of peptide receptor radiotherapy (PRRT) with radiolabeled beta radiation emitting somatostatin analogs (SSAs), several chemotherapy schedules (For a review see ref (75)) and targeted treatment with everolimus and sunitinib. PRRT with radiolabeled beta radiation emitting SSAs and, as mentioned above, mTOR inhibitors like everolimus, are frequently able to successfully control the hypoglycemias in patients with inoperable metastatic insulinomas (66-69, 75-79).

 

REFERENCES

 

  1. de Herder WW, Rehfeld JF, Kidd M, Modlin IM. A short history of neuroendocrine tumours and their peptide hormones. Best Pract Res Clin Endocrinol Metab. 2016;30(1):3-17.
  2. Mehrabi A, Fischer L, Hafezi M, Dirlewanger A, Grenacher L, Diener MK, et al. A systematic review of localization, surgical treatment options, and outcome of insulinoma. Pancreas. 2014;43(5):675-86.
  3. La Rosa S, Hong SM, Ohike N, Brosens LAA, Mete O, de Herder W. Insulinoma. In: Klimstra DS, Osamura RY, editors. WHO Classification of Tumours Endocrine and neuroendocrine tumours. 5 ed. Lyon: International Agency for Research on Cancer; 2023.
  4. Shames JM, Dhurandhar NR, Blackard WG. Insulin-secreting bronchial carcinoid tumor with widespread metastases. Am J Med. 1968;44(4):632-7.
  5. Adamson AR, Grahame-Smith DG, Bogomoletz V, Maw DS, Rothnie NG. Malignant argentaffinoma with carcinoid syndrome and hypoglycaemia. Br Med J. 1971;3(5766):93-4.
  6. Pelletier G, Cortot A, Launay JM, Debons-Guillemain MC, Nemeth J, Le Charpentier Y, et al. Serotonin-secreting and insulin-secreting ileal carcinoid tumor and the use of in vitro culture of tumoral cells. Cancer. 1984;54(2):319-22.
  7. La Rosa S, Pariani D, Calandra C, Marando A, Sessa F, Cortese F, et al. Ectopic duodenal insulinoma: a very rare and challenging tumor type. Description of a case and review of the literature. Endocr Pathol. 2013;24(4):213-9.
  8. Zhang X, Jia H, Li F, Fang C, Zhen J, He Q, et al. Ectopic insulinoma diagnosed by 68Ga-Exendin-4 PET/CT: A case report and review of literature. Medicine (Baltimore). 2021;100(13):e25076.
  9. Garg R, Memon S, Patil V, Bandgar T. Extrapancreatic insulinoma. World J Nucl Med. 2020;19(2):162-4.
  10. Rindi G, Mete O, Uccella S, Basturk O, La Rosa S, Brosens LAA, et al. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr Pathol. 2022;33(1):115-54.
  11. Hackeng WM, Brosens LAA, Dreijerink KMA. Aggressive versus indolent insulinomas - new clinicopathological insights. Endocr Relat Cancer. 2023.
  12. Svensson E, Muth A, Hedenström P, Ragnarsson O. The incidence of insulinoma in Western Sweden between 2002 and 2019. Ann Gastroenterol. 2022;35(4):434-40.
  13. Hofland J, Kaltsas G, de Herder WW. Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr Rev. 2020;41(2):371-403.
  14. de Herder WW, Niederle B, Scoazec JY, Pauwels S, Kloppel G, Falconi M, et al. Well-differentiated pancreatic tumor/carcinoma: insulinoma. Neuroendocrinology. 2006;84(3):183-8.
  15. Pieterman CRC, van Leeuwaarde RS, van den Broek MFM, van Nesselrooij BPM, Valk GD. Multiple Endocrine Neoplasia Type 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  16. Niederle B, Selberherr A, Bartsch D, Brandi ML, Doherty GM, Falconi M, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) and the Pancreas - Diagnosis and Treatment of Functioning and Non-Functioning Pancreatic and Duodenal Neuroendocrine Neoplasia within the MEN1 Syndrome - An International Consensus Statement. Neuroendocrinology. 2020.
  17. Alrezk R, Hannah-Shmouni F, Stratakis CA. MEN4 and CDKN1B mutations: the latest of the MEN syndromes. Endocr Relat Cancer. 2017;24(10):T195-t208.
  18. de Herder WW, Hofland J. Multiple Endocrine Neoplasia Type 4. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  19. Laks S, van Leeuwaarde R, Patel D, Keutgen XM, Hammel P, Nilubol N, et al. Management recommendations for pancreatic manifestations of von Hippel-Lindau disease. Cancer. 2022;128(3):435-46.
  20. Dworakowska D, Grossman AB. Are neuroendocrine tumours a feature of tuberous sclerosis? A systematic review. Endocr Relat Cancer. 2009;16(1):45-58.
  21. Service FJ. Hypoglycemic disorders. N Engl J Med. 1995;332(17):1144-52.
  22. Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, et al. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2009;94(3):709-28.
  23. Christ E, Iacovazzo D, Korbonits M, Perren A. Insulinomatosis - new aspects. Endocr Relat Cancer. 2023.
  24. Dauben L, Simon MC, Strassburger K, Burkart V, Weber KS, Schinner S, et al. Comparison of the diagnostic accuracy of the current guidelines for detecting insulinoma. Eur J Endocrinol. 2019;180(6):381-6.
  25. Le Roith D. Tumor-induced hypoglycemia. N Engl J Med. 1999;341(10):757-8.
  26. Iglesias P, Díez JJ. Management of endocrine disease: a clinical update on tumor-induced hypoglycemia. Eur J Endocrinol. 2014;170(4):R147-57.
  27. Iacovazzo D, Flanagan SE, Walker E, Quezado R, de Sousa Barros FA, Caswell R, et al. MAFA missense mutation causes familial insulinomatosis and diabetes mellitus. Proc Natl Acad Sci U S A. 2018;115(5):1027-32.
  28. Sempoux C, Kloppel G. Pathological features in non-neoplastic congenital and adult hyperinsulinism: from nesidioblastosis to current terminology and understanding. Endocr Relat Cancer. 2023.
  29. Bansal N, Weinstock RS. Non-Diabetic Hypoglycemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  30. Cimmino I, Faggiano A, Perruolo G, Modica R, Bottiglieri F, Covelli B, et al. Diagnosis of Flier's syndrome in a patient with nondiabetic hypoglycemia: a case report and critical appraisal of the literature. Endocrine. 2020;69(1):73-8.
  31. Oest L, Roden M, Müssig K. Comparison of patient characteristics between East Asian and non-East Asian patients with insulin autoimmune syndrome. Clin Endocrinol (Oxf). 2022;96(3):328-38.
  32. Ross JJ, Vaidya A, Kaiser UB. Interactive medical case. Lying low. N Engl J Med. 2011;364(6):e10.
  33. Vaidya A, Kaiser UB, Levy BD, Loscalzo J. Clinical problem-solving. Lying low. N Engl J Med. 2011;364(9):871-5.
  34. Pallais JC, Blake MA, Deshpande V. Case records of the Massachusetts General Hospital. Case 33-2012. A 34-year-old woman with episodic paresthesias and altered mental status after childbirth. N Engl J Med. 2012;367(17):1637-46.
  35. Wexler DJ, Macias-Konstantopoulos W, Forcione DG, Xiong L, Cauley CE, Pierce KJ. Case 23-2018: A 36-Year-Old Man with Episodes of Confusion and Hypoglycemia. N Engl J Med. 2018;379(4):376-85.
  36. Service FJ, Natt N. The prolonged fast. J Clin Endocrinol Metab. 2000;85(11):3973-4.
  37. Hofland J, Falconi M, Christ E, Castaño JP, Faggiano A, Lamarca A, et al. European Neuroendocrine Tumor Society (ENETS) 2023 Guidance Paper for Functioning Pancreatic Neuroendocrine Tumour Syndromes. J Neuroendocrinol. 2023.
  38. Druce MR, Muthuppalaniappan VM, O'Leary B, Chew SL, Drake WM, Monson JP, et al. Diagnosis and localisation of insulinoma: the value of modern magnetic resonance imaging in conjunction with calcium stimulation catheterisation. Eur J Endocrinol. 2010;162(5):971-8.
  39. de Herder WW. GEP-NETS update: functional localisation and scintigraphy in neuroendocrine tumours of the gastrointestinal tract and pancreas (GEP-NETs). Eur J Endocrinol. 2014;170(5):R173-83.
  40. Shah R, Sehemby M, Garg R, Purandare N, Hira P, Mahajan A, et al. (68) Ga-DOTATATE PET/CT imaging in endogenous hyperinsulinemic hypoglycemia: A tertiary endocrine centre experience. Clin Endocrinol (Oxf). 2022;96(2):190-9.
  41. Refardt J, Hofland J, Wild D, Christ E. Molecular Imaging of Neuroendocrine Neoplasms. J Clin Endocrinol Metab. 2022;107(7):e2662-e70.
  42. Shah R, Garg R, Majmundar M, Purandare N, Malhotra G, Patil V, et al. Exendin-4-based imaging in insulinoma localization: Systematic review and meta-analysis. Clin Endocrinol (Oxf). 2021;95(2):354-64.
  43. Boss M, Rottenburger C, Brenner W, Blankenstein O, Prasad V, Prasad S, et al. (68)Ga-NODAGA-Exendin-4 PET/CT Improves the Detection of Focal Congenital Hyperinsulinism. J Nucl Med. 2022;63(2):310-5.
  44. Imperiale A, Boursier C, Sahakian N, Ouvrard E, Chevalier E, Sebag F, et al. Value of (68)Ga-DOTATOC and Carbidopa-Assisted (18)F-DOPA PET/CT for Insulinoma Localization. J Nucl Med. 2022;63(3):384-8.
  45. Ismail D, Kapoor RR, Smith VV, Ashworth M, Blankenstein O, Pierro A, et al. The heterogeneity of focal forms of congenital hyperinsulinism. J Clin Endocrinol Metab. 2012;97(1):E94-9.
  46. Sada A, McKenzie TJ, Vella A, Levy MJ, Halfdanarson TR. Interventional versus surgical procedures in localized/nonmetastatic insulinoma (ablation versus surgery). Endocr Relat Cancer. 2023.
  47. de Mestier L, Hentic O, Cros J, Walter T, Roquin G, Brixi H, et al. Metachronous hormonal syndromes in patients with pancreatic neuroendocrine tumors: a case-series study. Ann Intern Med. 2015;162(10):682-9.
  48. Crona J, Norlén O, Antonodimitrakis P, Welin S, Stålberg P, Eriksson B. Multiple and Secondary Hormone Secretion in Patients With Metastatic Pancreatic Neuroendocrine Tumours. J Clin Endocrinol Metab. 2016;101(2):445-52.
  49. Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L, Buchler MW, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104(10):764-77.
  50. Rindi G, Klersy C, Albarello L, Baudin E, Bianchi A, Buchler MW, et al. Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: Data from a Large International Cohort Study. Neuroendocrinology. 2018;107(4):375-86.
  51. Hackeng WM, Schelhaas W, Morsink FHM, Heidsma CM, van Eeden S, Valk GD, et al. Alternative Lengthening of Telomeres and Differential Expression of Endocrine Transcription Factors Distinguish Metastatic and Non-metastatic Insulinomas. Endocr Pathol. 2020;31(2):108-18.
  52. Hofland J, de Herder WW, Kann PH. Turning Up the Heat: Endoscopic Ablation of Pancreatic Neuroendocrine Neoplasms. J Clin Endocrinol Metab. 2019;104(11):5053-5.
  53. Oleinikov K, Dancour A, Epshtein J, Benson A, Mazeh H, Tal I, et al. Endoscopic Ultrasound-Guided Radiofrequency Ablation: A New Therapeutic Approach for Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104(7):2637-47.
  54. Crinò SF, Napoleon B, Facciorusso A, Lakhtakia S, Borbath I, Caillol F, et al. Endoscopic Ultrasound-Guided Radiofrequency Ablation versus Surgical Resection for Treatment of Pancreatic Insulinoma. Clin Gastroenterol Hepatol. 2023.
  55. Habibollahi P, Bai HX, Sanampudi S, Soulen MC, Dagli M. Effectiveness of Liver-Directed Therapy for the Management of Intractable Hypoglycemia in Metastatic Insulinoma. Pancreas. 2020;49(6):763-7.
  56. Gill GV, Rauf O, MacFarlane IA. Diazoxide treatment for insulinoma: a national UK survey. Postgrad Med J. 1997;73(864):640-1.
  57. Ulbrecht JS, Schmeltz R, Aarons JH, Greene DA. Insulinoma in a 94-year-old woman: long-term therapy with verapamil. Diabetes Care. 1986;9(2):186-8.
  58. Taniguchi H, Murakami K, Morita S, Kazumi T, Yoshino G, Maeda M, et al. Calcium antagonist (diltiazem) for reversal of hypoglycaemic symptoms in insulinoma. Lancet. 1977;2(8036):501.
  59. Blum I, Aderka D, Doron M, Laron Z. Suppression of hypoglycemia by DL-propranolol in malignant insulinoma. N Engl J Med. 1978;299(9):487.
  60. Aderka D, Shaklai M, Doron M, Laron Z, Pinkhas J, De Vries A. Letter: Phenytoin in metastatic insulinoma. Jama. 1975;234(11):1119.
  61. Arnaout MA, Salti I. Letter: Phenytoin in benign insulinoma. Lancet. 1976;1(7964):861.
  62. Ito T, Lee L, Jensen RT. Treatment of symptomatic neuroendocrine tumor syndromes: recent advances and controversies. Expert Opin Pharmacother. 2016;17(16):2191-205.
  63. Husni H, Khan SA, Alghaieb B, Abusamaan MS, Donner TW, Hamrahian AH. Pasireotide use for the treatment of endogenous hyperinsulinemic hypoglycemia refractory to conventional medical therapy: A case report and review of the literature. Clin Case Rep. 2022;10(3):e05650.
  64. Oziel-Taieb S, Maniry-Quellier J, Chanez B, Poizat F, Ewald J, Niccoli P. Pasireotide for Refractory Hypoglycemia in Malignant Insulinoma- Case Report and Review of the Literature. Front Endocrinol (Lausanne). 2022;13:860614.
  65. Siddiqui M, Vora A, Ali S, Abramowitz J, Mirfakhraee S. Pasireotide: A Novel Treatment for Tumor-Induced Hypoglycemia Due to Insulinoma and Non-Islet Cell Tumor Hypoglycemia. J Endocr Soc. 2021;5(1):bvaa171.
  66. Yao JC, Pavel M, Lombard-Bohas C, Van Cutsem E, Voi M, Brandt U, et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers From the Randomized, Phase III RADIANT-3 Study. J Clin Oncol. 2016;34(32):3906-13.
  67. Bernard V, Lombard-Bohas C, Taquet MC, Caroli-Bosc FX, Ruszniewski P, Niccoli P, et al. Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia. Eur J Endocrinol. 2013;168(5):665-74.
  68. de Herder WW, van Schaik E, Kwekkeboom D, Feelders RA. New therapeutic options for metastatic malignant insulinomas. Clin Endocrinol (Oxf). 2011;75(3):277-84.
  69. Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195-7.
  70. Tovazzi V, Ferrari VD, Berruti A. Maintenance everolimus beyond progression in pancreatic NET to control insulinoma syndrome. Endocrine. 2021;71(1):258.
  71. Tovazzi V, Ferrari VD, Dalla Volta A, Consoli F, Amoroso V, Berruti A. Should everolimus be stopped after radiological progression in metastatic insulinoma? A "cons" point of view. Endocrine. 2020;69(3):481-4.
  72. Zandee WT, Hofland J, de Herder WW. Should everolimus be stopped after radiological progression in metastatic insulinoma? A "pro" point of view. Endocrine. 2021;71(1):256-7.
  73. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501-13.
  74. Service FJ. Recurrent hyperinsulinemic hypoglycemia caused by an insulin-secreting insulinoma. Nat Clin Pract Endocrinol Metab. 2006;2(8):467-70; quiz following 70.
  75. Garcia-Carbonero R, Anton-Pascual B, Modrego A, Riesco-Martinez MDC, Lens-Pardo A, Carretero-Puche C, et al. Advances in the Treatment of Gastroenteropancreatic Neuroendocrine Carcinomas: are we moving forward? Endocr Rev. 2023.
  76. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514-23.
  77. Zandee WT, Brabander T, Blazevic A, Kam BLR, Teunissen JJM, Feelders RA, et al. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104(4):1336-44.
  78. van Schaik E, van Vliet EI, Feelders RA, Krenning EP, Khan S, Kamp K, et al. Improved control of severe hypoglycemia in patients with malignant insulinomas by peptide receptor radionuclide therapy. J Clin Endocrinol Metab. 2011;96(11):3381-9.
  79. Verma P, Malhotra G, Dodamani MH, Lila AR, Asopa RV, Bandgar TR. Complete Resolution of Disease After Peptide Receptor Radionuclide Therapy in a Patient of Metastatic Insulinoma. Clin Nucl Med. 2022;47(1):e77-e8.

 

Somatostatinoma

ABSTRACT

 

Somatostatin-secreting tumors, or somatostatinomas represent less than 1% of functioning gastrointestinal neuroendocrine neoplasms (NENs) and their estimated incidence is about 1 in 40 million individuals per year. The spectrum of the somatostatinoma syndrome consists of diabetes mellitus, diarrhea/steatorrhea, cholelithiasis, hypochlorhydria, and weight loss. Tumors that demonstrate D-cell differentiation based on immunohistochemical labelling with somatostatin but lack symptoms of somatostatinoma syndrome, such as those observed within the ampulla and duodenum, should be designated as somatostatin-producing well-differentiated NENs and are not considered somatostatinomas. Hereditary pancreatic somatostatin-producing well-differentiated NENs can be found as part of multiple neuroendocrine neoplasia type 1 (MEN1) and von-Hippel Lindau (VHL) syndrome, whereas duodenal (peri-ampullary somatostatin-producing NENs can be found in patients with neurofibromatosis type 1 (NF1). The polycythemia-paraganglioma-somatostatinoma syndrome is a rare syndrome including multiple paragangliomas, duodenal somatostatin-producing NENs (exclusively found at the ampulla of Vater) associated with high erythropoietin (polycythemia) underlying paraganglioma/pheochromocytoma. The diagnosis of a somatostatinoma requires measuring fasting plasma somatostatin hormone concentration. A 3-phase CT, MRI, positron emission tomography (PET)-CT with gallium-labelled somatostatin analogs, or endoscopic ultrasound (EUS) should be performed for the precise localization of somatostatinomas in the pancreas or duodenum. A biopsy or surgical resection is required for grading (Ki67 index) and immunohistochemistry for somatostatin expression on tumor samples. Management of somatostatinomas includes medical treatment of the excess somatostatin production, surgical and/or radiological interventions, peptide receptor radiotherapy and targeted or cytotoxic therapies.

 

INTRODUCTION

 

The tetradecapeptide somatostatin is the main peptide released from somatostatinomas. This hormone was successfully isolated in 1973 by Paul Brazeau and colleagues in the research group of the French-US endocrinologist and Nobel prize laureate Roger Guillemin (1). Somatostatin inhibits numerous endocrine and exocrine secretory functions. Almost all gut hormones are inhibited by somatostatin, including insulin, glucagon, gastrin, secretin, and gastric inhibitory polypeptide (GIP). In addition to inhibition of the endocrine secretions, somatostatin has direct effects on a number of other target organs. For example, it is an inhibitor of gastric acid and pancreatic enzyme secretion, it has marked effects on gastrointestinal transit time, intestinal motility, and absorption of nutrients from the small intestine. In the nervous system, somatostatin acts as a neurotransmitter or neuromodulator and its roles in the fine-tuning of neuronal activity and involvement in synaptic plasticity and memory formation are now widely recognized (2).

 

In 1977, the groups of the Danish physician Lars-Inge Larsson and that of the US physician Om P. Ganda independently reported the first two cases of pancreatic somatostatinoma (3, 4). In 1979, a full description of the somatostatinoma syndrome caused by a periampullary neuroendocrine tumor was reported by the Austrian gastroenterologist Günter Krejs and colleagues (5, 6). Since then, numerous cases have been reported until present, most of them being sporadic, but some of them now being recognized as part of classic or emerging genetic syndromes.

 

Somatostatinomas are very rare neuroendocrine neoplasms (NENs); their estimated incidence is about 1 in 40 million individuals per year in the general population and they account for less than 1% of functioning pancreatic neuroendocrine neoplasms (PanNENs). These panNENs can arise throughout the pancreas, but approximately two thirds involve the head of this organ (7).The mean patient age at diagnosis is 55 years and these tumors occur more commonly in females (7-9).

 

CLINICAL PRESENTATION

 

Although somatostatinomas secrete somatostatin, clinical presentations related to high somatostatin levels are found in less than 5% of cases. This may depend on the location of the NEN (generally pancreatic), as well as intermittent somatostatin secretion from the NEN  (3, 9). The spectrum of the somatostatinoma syndrome consists of diabetes mellitus, diarrhea/steatorrhea, cholelithiasis, hypochlorhydria, and weight loss. This implies that the majority of somatostatinomas do not present with the typical somatostatinoma symptoms, but are silent. Tumors that demonstrate D-cell differentiation based on immunohistochemical labelling with somatostatin but lack symptoms of somatostatinoma syndrome, such as those observed within the ampulla and duodenum, should be designated as somatostatin-producing well-differentiated NENs and are not considered somatostatinomas. Patients harboring these tumors experience symptoms related to the tumor mass effect, their metastases, or the invasion of contiguous structures. Therefore, these silent NENs are generally detected by Computed Tomography (CT), Magnetic Resonance Imaging (MRI) or, on occasion, by Somatostatin Receptor Imaging (SRI) and Endoscopic Ultrasonography (EUS). The most common symptom for all somatostatin-producing well-differentiated NENs is abdominal pain, occurring in over 50% of patients. Duodenal tumors can also present with jaundice and gastrointestinal bleeding (9-11).

 

Secretion of different hormones by the same panNEN, sometimes resulting in two, or more synchronous, or metachronous distinct endocrine syndromes, is now being recognized with increasing frequency. However, second, or metachronous somatostatin secretion has thus far not been recognized. These possibilities should be considered during endocrine work-up and follow-up of patients with panNENs (12, 13).

 

Somatostatin has been found in many tissues outside the GI tract. Prominent among those are the hypothalamic and extrahypothalamic regions of the brain, the peripheral nervous system (including the sympathetic adrenergic ganglia), and the C cells of the thyroid gland. Therefore, high plasma concentrations of somatostatin have been found in tumors originating from these tissues (14). Pheochromocytomas and paragangliomas are other examples of neuroendocrine tumors that produce and secrete somatostatin in addition to other hormonally active substances (15). However, these tumors do not present with signs or symptoms of the somatostatinoma syndrome.

 

HEREDITARY TUMORS

 

Hereditary pancreatic somatostatin-producing NENs can be found as part of multiple neuroendocrine neoplasia type 1 (MEN1) and von-Hippel Lindau (VHL) syndrome, whereas duodenal, peri-ampullary, somatostatin-producing NENs can be found in patients with neurofibromatosis type 1 (NF1) (16-20). An overview of the MEN1 syndrome is provided in the chapter “MEN1”. Previously known as Von Recklinghausen disease, NF1, the most frequent neuro-cutaneous syndrome, is an autosomal dominant condition. The reported incidence is 1/2500-1/3000 (39,40). The mutation causing the condition is at the level of NF1 gene (on chromosome 17) which induces a malfunction of the RAS/MAPK pathway. The presence of a duodenal somatostatin-producing NEN has a higher risk in NF1 patients than in the general population, but this is not the most prevalent tumor encountered in these patients. Some studies report on the combined diagnosis of GIST and somatostatin-producing NENs in subjects with neurofibromatosis type 1 (9-11, 21-25).

 

The polycythemia-paraganglioma-somatostatinoma syndrome (also called Pacak-Zhuang syndrome) is a rare new syndrome including multiple paragangliomas, duodenal somatostatin-producing NENs, which are exclusively found in the region of the ampulla of Vater, and a high circulating erythropoietin concentration resulting in polycythemia. A gain of function involving the mutation of Endothelial PAS domain-containing protein 1 [EPAS1, also known as hypoxia-inducible factor-2alpha (HIF-2alpha)] gene underlies the Pacak-Zhuang syndrome. Moreover, non-mosaicism somatic mutations of HIF-2alpha seem to induce the same syndrome but with late onset. A somatic gain-of-function HIF-2alpha mutation results in the stabilization of HIF-2α, which is known to upregulate the erythropoietin gene accounting for polycythemia in these patients (25-33).

 

However, while the association of somatostatinomas / somatostatin-producing NENs with these inherited disorders is intriguing, a link between the known gene mutations of these disorders with the development of somatostatin-producing NENs has not been clearly established.

 

DIAGNOSIS

 

The diagnosis of somatostatinoma requires the combination of typical clinical signs and symptoms with measuring the fasting plasma somatostatin hormone concentration, which should be at least 3 times over the upper reference value (> 25 pmol/L (> 60 pg/mL). In case of an indeterminate test result, stimulatory examinations such as secretin or calcium stimulation tests can be used, but these tests lack standardization. Anatomic and functional imaging modalities are important in the localization of a somatostatinoma. As in other NENs, 3-phase CT, MRI, or endoscopic ultrasound (EUS) should be performed for the precise localization of these tumors in the pancreas or duodenum. To detect distant metastases, somatostatin receptor imaging should be used as somatostatinomas express high numbers of different somatostatin receptor subtypes. Currently, positron emission tomography (PET)-CT with 68Ga-labelled somatostatin analogs (DOTATATE, DOTANOC, DOTATOC) has the highest sensitivity for detecting metastases of grade 1-3 panNENs (34, 35). In line with the work-up for all NENs, a biopsy is advised to confirm the diagnosis and for grading (Ki67 index), as the grade can influence treatment selection. An overview of the current staging and grading systems is provided in the chapter “Insulinoma” (36). Generally, though, pathological examination and immunohistochemistry for somatostatin expression on tumor samples after surgery or biopsy confirms the definitive diagnosis. The tumor further shows diffuse positivity for keratins, INSM1 and synaptophysin and is less consistently positive or negative for chromogranin A (7, 9, 35, 37), see Figure 1.

Figure 1.  A malignant metastasizing pancreatic endocrine tumor located in the tail of the pancreas with positive immunohistochemical staining of somatostatin (Courtesy of Günther Klöppel).

 

TREATMENT

 

The management of somatostatinomas includes medical treatment of the excess somatostatin production, surgical and/or radiological interventions, and cytotoxic therapies when needed (35).

 

Curative Surgery

 

As for all panNENs, surgery is the only curative treatment. In the occasional patient in whom a somatostatinoma is discovered while the tumor is locoregionally confined, pancreatic or duodenal surgery should be performed to remove the somatostatinoma. In selected patients with limited liver metastases an extended surgical resection can be considered (38).

 

Liver-Directed Therapy

 

Liver metastases can be resected or treated by bland embolization, radioembolization (SIRT), radiofrequency ablation (RFA), microwave and cryoablation, high-intensity focused ultrasound (HIFU), laser, brachytherapy and irreversible electroporation (IRE) depending on local availability (39, 40).

 

Unresectable Disease

 

In case of unresectable metastases, treatment is focused on tumor stabilization and symptom reduction by decreasing the secretion of somatostatin. In general, anti-tumor therapy is similar to non-functioning panNENs as specific data for somatostatinoma are often lacking. The guidelines by ENETS, NANETS and ESMO describe the selection and sequencing of somatostatin analogs, targeted therapy, 177Lu-DOTATATE and cytotoxic chemotherapy (41-44).

 

SOMATOSTATIN ANALOGS

 

Somatostatin analogs became an important treatment option for patients with metastatic or inoperable NENs. First, these analogs provide relief of symptoms in patients with NENs that secrete different peptides causing various clinical symptoms and signs, especially diarrhea and weight loss in somatostatinoma patients (45). Somatostatin analogs are the first-line palliative treatment of choice to control somatostatin secretion and tumor growth. In a randomized controlled trial (CLARINET), including grade 1-2 pancreatic neuroendocrine tumors (NETs - panNETs), lanreotide autogel 120 mg every 4 weeks deep sc was associated with significantly prolonged median progression-free survival (PFS) of 38 months versus 18 months for placebo (46).

 

BELZUTIFAN

 

In the polycythemia-paraganglioma-somatostatinoma syndrome (Pacak-Zhuang syndrome) treatment with the HIF-2alpha inhibitor Belzutifan resulted in a reduction/normalization of the pathologically elevated levels of normetanephrine, Chromogranin A, Hemoglobin and Erythropoietin (47). Up to present no effects of this drug have been described on the somatostatin-producing NETs in these patients. Belzutifan is also used for the treatment of tumors in patients with the VHL syndrome (48, 49).

 

PEPTIDE RECEPTOR RADIONUCLIDE THERAPY

 

The expression of somatostatin receptor subtypes provides an opportunity to utilize peptide receptor radionuclide therapy (PRRT) for the treatment of metastatic somatostatinomas. PRRT with 177Lu-DOTATATE has been approved for the treatment of grade 1-2 panNETs. In general, the response rate for grade 1-2 panNETs is the highest of all NETs (55%), with a median progression-free survival (PFS) of 30 months and median overall survival (OS) of 71 months. Sub-acute toxicity mainly includes nausea, vomiting, and criteria for adverse events (CTCAE) grade 3/4 toxicity of hematologic parameters (10%). In 70% of patients with toxicity, the hematologic parameters normalize but 1% of patients treated with PRRT develops acute leukemia, and 2% myelodysplastic syndrome (50). In patients with uncontrollable hypersecretion by hormone-producing panNENs, PRRT with 177Lu-DOTATATE can result in amelioration of the hormonal syndrome (51). However, data of PRRT with 177Lu-DOTATATE for the treatment of metastatic somatostatinoma are not available yet.

 

EVEROLIMUS

 

Everolimus is an oral drug which inhibits mammalian target of rapamycin (mTOR) signaling. In the RADIANT-3 trial, everolimus 10 mg/day increased progression-free survival in grade 1-2 panNETs to 11.0 months as compared to 4.6 months with placebo. Also, overall survival did increase from 37.6 to 44 months. In this study 24% of patients had a functioning grade 1-2 panNET including somatostatinoma (52). As everolimus can also worsen diabetes mellitus by reducing insulin secretion from the pancreas and inducing insulin resistance, its contribution to the treatment of somatostatinoma patients is still unclear.

 

SUNITINIB

 

Sunitinib is currently one of the other options for treatment of grade 1-2 panNETs which progress during treatment with a first generation long-acting somatostatin analog. In the SU011248 trial sunitinib 37.5 mg/day increased progression-free survival to 11.4 months in comparison to 5.5 months with placebo in patients with predominantly grade 1-2 panNETs. Overall survival also increased from 29.1 to 38.6 months. In this trial, only one patient with a somatostatinoma was included in the treatment arm (53, 54).

 

CHEMOTHERAPY

 

Chemotherapy is also effective for the treatment of panNEN but no specific data for somatostatinoma are available (42, 55, 56).

 

REFERENCES

 

  1. de Herder WW, Rehfeld JF, Kidd M, Modlin IM. A short history of neuroendocrine tumours and their peptide hormones. Best Pract Res Clin Endocrinol Metab. 2016;30(1):3-17.
  2. Schubert ML, Rehfeld JF. Gastric Peptides-Gastrin and Somatostatin. Compr Physiol. 2019;10(1):197-228.
  3. Larsson LI, Hirsch MA, Holst JJ, Ingemansson S, Kühl C, Jensen SL, et al. Pancreatic somatostatinoma. Clinical features and physiological implications. Lancet. 1977;1(8013):666-8.
  4. Ganda OP, Weir GC, Soeldner JS, Legg MA, Chick WL, Patel YC, et al. "Somatostatinoma": a somatostatin-containing tumor of the endocrine pancreas. N Engl J Med. 1977;296(17):963-7.
  5. Krejs GJ, Collins SM, McCarthy D, Unger RH, Gardner JD. Follow-up of a patient with somatostatinoma. N Engl J Med. 1986;315(20):1295.
  6. Krejs GJ, Orci L, Conlon JM, Ravazzola M, Davis GR, Raskin P, et al. Somatostatinoma syndrome. Biochemical, morphologic and clinical features. N Engl J Med. 1979;301(6):285-92.
  7. Brosens LA, Singhi AD, Basturk O, Hong SM, La Rosa S, Mete O, et al. Somatostatinoma. In: Klimstra DS, Osamura RY, editors. WHO Classification of Tumours Endocrine and neuroendocrine tumours. Lyon: International Agency for Research on Cancer; 2023.
  8. Mastoraki A, Schizas D, Papoutsi E, Ntella V, Kanavidis P, Sioulas A, et al. Clinicopathological Data and Treatment Modalities for Pancreatic Somatostatinomas. In Vivo. 2020;34(6):3573-82.
  9. Garbrecht N, Anlauf M, Schmitt A, Henopp T, Sipos B, Raffel A, et al. Somatostatin-producing neuroendocrine tumors of the duodenum and pancreas: incidence, types, biological behavior, association with inherited syndromes, and functional activity. Endocr Relat Cancer. 2008;15(1):229-41.
  10. Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Case 15-1989. A 52-year-old man with neurofibromatosis and jaundice. N Engl J Med. 1989;320(15):996-1004.
  11. Thavaraputta S, Graham S, Rivas Mejia AM, Lado-Abeal J. Duodenal somatostatinoma presenting as obstructive jaundice with the coexistence of a gastrointestinal stromal tumour in neurofibromatosis type 1: a case with review of the literature. BMJ Case Rep. 2019;12(1).
  12. Crona J, Norlén O, Antonodimitrakis P, Welin S, Stålberg P, Eriksson B. Multiple and Secondary Hormone Secretion in Patients With Metastatic Pancreatic Neuroendocrine Tumours. J Clin Endocrinol Metab. 2016;101(2):445-52.
  13. de Mestier L, Hentic O, Cros J, Walter T, Roquin G, Brixi H, et al. Metachronous hormonal syndromes in patients with pancreatic neuroendocrine tumors: a case-series study. Ann Intern Med. 2015;162(10):682-9.
  14. Roos BA, Lindall AW, Ells J, Elde R, Lambert PW, Birnbaum RS. Increased plasma and tumor somatostatin-like immunoreactivity in medullary thyroid carcinoma and small cell lung cancer. J Clin Endocrinol Metab. 1981;52(2):187-94.
  15. Berelowitz M, Szabo M, Barowsky HW, Arbel ER, Frohman LA. Somatostatin-like immunoactivity and biological activity is present in a human pheochromocytoma. J Clin Endocrinol Metab. 1983;56(1):134-8.
  16. Niederle B, Selberherr A, Bartsch D, Brandi ML, Doherty GM, Falconi M, et al. Multiple Endocrine Neoplasia Type 1 (MEN1) and the Pancreas - Diagnosis and Treatment of Functioning and Non-Functioning Pancreatic and Duodenal Neuroendocrine Neoplasia within the MEN1 Syndrome - An International Consensus Statement. Neuroendocrinology. 2020.
  17. Hammel PR, Vilgrain V, Terris B, Penfornis A, Sauvanet A, Correas JM, et al. Pancreatic involvement in von Hippel-Lindau disease. The Groupe Francophone d'Etude de la Maladie de von Hippel-Lindau. Gastroenterology. 2000;119(4):1087-95.
  18. Maki M, Kaneko Y, Ohta Y, Nakamura T, Machinami R, Kurokawa K. Somatostatinoma of the pancreas associated with von Hippel-Lindau disease. Intern Med. 1995;34(7):661-5.
  19. Karasawa Y, Sakaguchi M, Minami S, Kitano K, Kawa S, Aoki Y, et al. Duodenal somatostatinoma and erythrocytosis in a patient with von Hippel-Lindau disease type 2A. Intern Med. 2001;40(1):38-43.
  20. Lévy-Bohbot N, Merle C, Goudet P, Delemer B, Calender A, Jolly D, et al. Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterol Clin Biol. 2004;28(11):1075-81.
  21. Deschamps L, Dokmak S, Guedj N, Ruszniewski P, Sauvanet A, Couvelard A. Mixed endocrine somatostatinoma of the ampulla of vater associated with a neurofibromatosis type 1: a case report and review of the literature. Jop. 2010;11(1):64-8.
  22. Kumar T, Gupta B, Das P, Jain D, Jain HA, Madhusudhan KS, et al. Combined presence of multiple gastrointestinal stromal tumors along with duodenal submucosal somatostatinoma in a patient with neurofibromatosis type 1. Indian J Pathol Microbiol. 2016;59(3):359-61.
  23. Luna IE, Monrad N, Binderup T, Boisen Thoegersen C, Hilsted L, Jensen C, et al. Somatostatin-Immunoreactive Pancreaticoduodenal Neuroendocrine Neoplasms: Twenty-Three Cases Evaluated according to the WHO 2010 Classification. Neuroendocrinology. 2016;103(5):567-77.
  24. Martin S, Fica S, Parfeni O, Popa L, Manuc T, Rizea O, et al. Somatostatinoma and Neurofibromatosis Type 1-A Case Report and Review of the Literature. Diagnostics (Basel). 2020;10(9).
  25. Sandru F, Carsote M, Valea A, Albu SE, Petca RC, Dumitrascu MC. Somatostatinoma: Beyond neurofibromatosis type 1 (Review). Exp Ther Med. 2020;20(4):3383-8.
  26. Därr R, Nambuba J, Del Rivero J, Janssen I, Merino M, Todorovic M, et al. Novel insights into the polycythemia-paraganglioma-somatostatinoma syndrome. Endocr Relat Cancer. 2016;23(12):899-908.
  27. Dwight T, Kim E, Bastard K, Benn DE, Eisenhofer G, Richter S, et al. Functional significance of germline EPAS1 variants. Endocr Relat Cancer. 2020.
  28. Pacak K, Jochmanova I, Prodanov T, Yang C, Merino MJ, Fojo T, et al. New syndrome of paraganglioma and somatostatinoma associated with polycythemia. J Clin Oncol. 2013;31(13):1690-8.
  29. Pang Y, Gupta G, Jha A, Yue X, Wang H, Huynh TT, et al. Nonmosaic somatic HIF2A mutations associated with late onset polycythemia-paraganglioma syndrome: Newly recognized subclass of polycythemia-paraganglioma syndrome. Cancer. 2019;125(8):1258-66.
  30. Tella SH, Taïeb D, Pacak K. HIF-2alpha: Achilles' heel of pseudohypoxic subtype paraganglioma and other related conditions. Eur J Cancer. 2017;86:1-4.
  31. Yang C, Hong CS, Prchal JT, Balint MT, Pacak K, Zhuang Z. Somatic mosaicism of EPAS1 mutations in the syndrome of paraganglioma and somatostatinoma associated with polycythemia. Hum Genome Var. 2015;2:15053.
  32. Yang C, Sun MG, Matro J, Huynh TT, Rahimpour S, Prchal JT, et al. Novel HIF2A mutations disrupt oxygen sensing, leading to polycythemia, paragangliomas, and somatostatinomas. Blood. 2013;121(13):2563-6.
  33. Zhuang Z, Yang C, Lorenzo F, Merino M, Fojo T, Kebebew E, et al. Somatic HIF2A gain-of-function mutations in paraganglioma with polycythemia. N Engl J Med. 2012;367(10):922-30.
  34. Refardt J, Hofland J, Wild D, Christ E. Molecular Imaging of Neuroendocrine Neoplasms. J Clin Endocrinol Metab. 2022;107(7):e2662-e70.
  35. Hofland J, Kaltsas G, de Herder WW. Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr Rev. 2020;41(2):371-403.
  36. de Herder WW, Hofland J. Insulinoma. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  37. Rindi G, Klersy C, Albarello L, Baudin E, Bianchi A, Buchler MW, et al. Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: Data from a Large International Cohort Study. Neuroendocrinology. 2018;107(4):375-86.
  38. Partelli S, Bartsch DK, Capdevila J, Chen J, Knigge U, Niederle B, et al. ENETS Consensus Guidelines for Standard of Care in Neuroendocrine Tumours: Surgery for Small Intestinal and Pancreatic Neuroendocrine Tumours. Neuroendocrinology. 2017;105(3):255-65.
  39. Frilling A, Al-Nahhas A, Clift AK. Transplantation and Debulking Procedures for Neuroendocrine Tumors. Front Horm Res. 2015;44:164-76.
  40. Frilling A, Clift AK. Surgical Approaches to the Management of Neuroendocrine Liver Metastases. Endocrinol Metab Clin North Am. 2018;47(3):627-43.
  41. Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(7):844-60.
  42. Pavel M, Valle JW, Eriksson B, Rinke A, Caplin M, Chen J, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasms: Systemic Therapy - Biotherapy and Novel Targeted Agents. Neuroendocrinology. 2017;105(3):266-80.
  43. Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, et al. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology. 2016;103(2):153-71.
  44. Halfdanarson TR, Strosberg JR, Tang L, Bellizzi AM, Bergsland EK, OʼDorisio TM, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(7):863-81.
  45. Angeletti S, Corleto VD, Schillaci O, Marignani M, Annibale B, Moretti A, et al. Use of the somatostatin analogue octreotide to localise and manage somatostatin-producing tumours. Gut. 1998;42(6):792-4.
  46. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224-33.
  47. Kamihara J, Hamilton KV, Pollard JA, Clinton CM, Madden JA, Lin J, et al. Belzutifan, a Potent HIF2α Inhibitor, in the Pacak-Zhuang Syndrome. N Engl J Med. 2021;385(22):2059-65.
  48. Pelle E, Al-Toubah T, Morse B, Strosberg J. Belzutifan in a Patient With VHL-Associated Metastatic Pancreatic Neuroendocrine Tumor. J Natl Compr Canc Netw. 2022;20(12):1285-7.
  49. Shepherd STC, Drake WM, Turajlic S. The road to systemic therapy in von Hippel-Lindau (VHL) disease: Are we there yet? Eur J Cancer. 2023;182:15-22.
  50. Brabander T, van der Zwan WA, Teunissen JJM, Kam BLR, Feelders RA, de Herder WW, et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin Cancer Res. 2017;23(16):4617-24.
  51. Zandee WT, Brabander T, Blazevic A, Kam BLR, Teunissen JJM, Feelders RA, et al. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104(4):1336-44.
  52. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):514-23.
  53. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501-13.
  54. Herrera-Martinez AD, Hofland J, Hofland LJ, Brabander T, Eskens F, Galvez Moreno MA, et al. Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives. Drugs. 2019;79(1):21-42.
  55. Garcia-Carbonero R, Rinke A, Valle JW, Fazio N, Caplin M, Gorbounova V, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasms. Systemic Therapy 2: Chemotherapy. Neuroendocrinology. 2017;105(3):281-94.
  56. Garcia-Carbonero R, Anton-Pascual B, Modrego A, Riesco-Martinez MDC, Lens-Pardo A, Carretero-Puche C, et al. Advances in the Treatment of Gastroenteropancreatic Neuroendocrine Carcinomas: are we moving forward? Endocr Rev. 2023.

 

 

 

 

Functional Anatomy of the Hypothalamus and Pituitary

ABSTRACT

 

In mammals and man, historical investigation suggests that early recognition for a role of the hypothalamus as a site for integration of endocrine, autonomic and behavioral responses can be dated to the 2nd -18th centuries A.D. Although the hypothalamus comprises only 2% of the total brain volume, it is a key regulator of pituitary function and homeostatic balance. In this chapter, we provide an overview of the historical landmarks, embryologic, gross, microscopic and functional anatomy of the mammalian and human hypothalamus and pituitary, and how the hypothalamus relates to the rest of the brain and responds to peripheral signals. In particular, we show that its rostral, nuclear portion exerts prominent regulation of homeostatic behaviors related to energy balance and reproduction. The two caudal portions are primarily involved in ensuring adequate metabolic resources for defensive and exploratory behaviors and responses to sudden changes in endogenous and exogenous stimuli. In addition, we discuss how its network of neurons is made of cells with different functions (neurosecretory, autonomic, motor), how they interact, and how these neural circuitries are woven into a complex architecture of conduits for the movement of intercellular fluids (vasculature, glymphatic channels, meningeal lymphatic vessels). Finally, we focus on the hypothalamic mechanisms involved in the regulation of anterior and posterior pituitary secretion (hypothalamic tuberoinfundibular and neurohypophysial systems), as well those involved in food and fluid intake, lactation, thermoregulation, circadian rhythmicity and the sleep-wake cycle.

 

HISTORICAL OVERVIEW

 

As suggested by its Greek derivation, the hypothalamus (hypo = below, thalamus = bed) is that portion of the diencephalon in all vertebrates that lies inferior to the thalamus (1).  The hypothalamus and pituitary gland have attracted the interest of scientists and artists for centuries since the first description by Galen of Pergamon in the 2nd century AD.  Galen described the hypothalamic infundibulum and the pituitary gland in De Usu Partium as the draining route and receptacle, respectively, for mucus passing from the brain ventricular structures (primarily the third ventricle) to the nasopharynx, and named the capillary network surrounding the pituitary gland the rete mirabilis (2). Notably, he also recognized the association of the third ventricle with a dorsally-located, small gland he named “pineal”. The Galenic concepts dominated scientific thought about the hypothalamus and pituitary for approximately 1200 years until the 14th century when the Italian anatomist, Mondino de’ Liuzzi, in his Anothomia, proposed that the third ventricle serves as an “integrator” of body functions (Fig. 1) (3).  Some of these ideas were extended by Andreas Vesalius in the 16th century in De Humani Corporis Fabrica, the first anatomical depiction of the infudibular-pituitary stalk including part of their venous drainage, consistent with our current anatomical knowledge for petrosal sinus sampling (Fig. 2).  Attention to the importance of the hypothalamic-pituitary region influenced the work of some of the most famous Renaissance artists including Leonardo da Vinci, whose drawings of the third ventricle and rete mirabilis are shown in Fig. 3, and Michelangelo Buonarroti, whose painting on the ceiling of the Sistine Chapel in the Vatican uses the brain including the hypothalamic-pituitary region as a backdrop to his depiction of the creation of man (Fig. 4) (4).  Further interest in the functional role of the third ventricle occurred during the 17th century by the philosopher, Renè Descartes.  He hypothesized that a photic stimulus might reach the pineal gland from the retina, passing through the optic chiasm and third ventricle to stimulate the somatic motor nerves destined to the peripheral muscles to produce movement (Fig. 5).

 

Figure. 1. Description of the functional role exerted by the cerebral third ventricle, as reported by Mondino de’ Liuzzi in Anothomia. (A) Original front page of Anothomia in a XIV century edition; (B) Original text (in brackets) in medieval Latin (from the 1316 A.D. manuscript kept at the Società Medica Chirurgica in Bologna, Italy); (C) a portion of the Latin fragment shown in (B) containing the most important concepts; (D) English translation shown in (B). (From Toni R., Ancient views on the hypothalamic-pituitary-thyroid axis: an historical and epistemological perspective, Pituitary 3: 83-95, 2000).

Figure 2. Plates from the seventh book of the first edition (1543) of the Fabrica by Andreas Vesalius, showing what is believed to be the oldest anatomical drawings in Western literature of the hypothalamic-pituitary unit. (Courtesy of the Library of the Department of Human Anatomy of the University of Bologna, Italy, with permission) 1) Enlarged view of the pituitary gland (A), hypothalamic infundibulum (B) and ducts comprising the foramen lacerum and superior orbital fissure (C, D, E, F) believed to drain brain mucus or phlegm (in Latin pituita) from the pituitary gland to the nasopharynx; 2) anatomical relationships between the infundibulum (E), the dural diaphragma sellae (F), the internal carotid arteries (C, D) and oculomotor nerves (G), all seen from above and, thus ventral to the posterior clinoid processes of the sella turcica (A, B); 3) composite image including a) an enlarged view of the rete mirabilis formed as a reticular plexus by the carotid arteries entering (A, B) and emerging (C, D) around the pituitary gland (E); b) detailed view of the reticular plexus arising from the carotids (B, C) on each side of the pituitary (A); 4) anatomy of the arterial, vertebral (dorsal vessels, F) and common carotid (ventral vessels, E) systems: the rete mirabilis (B) is provided by the internal carotid artery (D), branching medially with respect to the external carotid artery (C). Note that Vesalius portrayed the rete mirabilis widening symmetrically and superiorly (A) to vascularize the area of the infundibulum and hypothalamic floor, anticipating our current knowledge of the circuminfundibular and prechiasmal arteriolar-capillary plexus; 5) anatomy of the venous vertebral (D) and internal jugular (C) systems, including the common facial vein (D). Note the X-shaped, venous pattern at the center of the image, pointing to the area of the rete mirabilis: it is provided by four symmetrical branches of the internal jugular vein, and recapitulates the distribution of the inferior and superior petrosal, and spheno-parietal sinuses around the cavernous sinus. Thus, this drawing can be considered the first demonstration of a venous route from the pituitary through the internal jugular system, exploited for sampling of pituitary hormonal secretions only in the 2nd half of the 20th century. (From Toni R., Ancient views on the hypothalamic-pituitary-thyroid axis: an historical and epistemological perspective, Pituitary 3: 83-95, 2000, and Toni R. “Il sistema ipotalamo-ipofisario nell’antichità [The hypothalamic-pituitary system in the antiquity] - Dedicato alla memoria del Prof. Aldo Pinchera [Dedicated to the memory of Prof. Aldo Pinchera], In: L’Endocrinologo, Per una Storia dell’Endocrinologia [For a History of Endocrinology], 13, suppl. to n. 6, 1-11, 2012.

Figure 3. Drawings by Leonardo da Vinci (1508-1509) taken from the Codici di Anatomia of the Windsor’s Collection (Courtesy of the Library of the Department of Human Anatomy of the University of Parma, Italy). (A) Inferior surface of the brain, showing the rete mirabilis (arrow) that surrounds the pituitary gland; (B) three-dimensional representation of the cerebral ventricles. The third ventricle (3v) was believed to be the site of afference and elaboration of the “sensus communis” (Latin for peripheral physical sensations). (From Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.)

Figure 4. Detail from the fresco, “Creation of Adam,” by Michelangelo Buonarroti, visible on the ceiling of the Sistine Chapel in the Vatican at Rome, Italy, painted between 1508-1512. (A) Photograph of the fresco showing God giving spiritual life and intellect to Adam through his touch; (B) The contour of the same image is reminiscent of a midline sagittal section of the brain and includes the hypothalamus, pituitary and brainstem. (From Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.)

Figure 5. Drawing from the De Homine of Descartes (1662), showing the pathway of the light through the ocular globe, retina, and collaterals of the optic nerves (corresponding to the retino-hypothalamic tract) that project to the 3rd ventricle (i.e., to the suprachiasmatic nucleus – SCN), to stimulate the pineal gland to release the animal spirit (corresponding to the nerve impulse) to the peripheral striatal muscles. Indeed, we currently know that the photic stimuli may not only activate a hypothalamic-medullary-epithalamic pathway for melatonin release (see Circadian Rhythm section) and thus signal the day/night shift to SCN-dependent pituitary secretions, but also trigger the lateral habenular nucleus. The lateral habenular nucleus receives projections from the SCN directly through the stria medullaris of the thalamus, and indirectly via the superior colliculi by lateral hypothalamic efferents. It influences brainstem motor centers like the substantia nigra and reticular raphe nuclei, to regulate body movement in relation to visual clues. In addition, melatonin acts on skeletal muscle as an ergogenic factor, favoring aerobic motor performance. Thus, Descartes view of the hypothalamic-pineal connection is partly consistent with evidence that light impulses transmitted through the hypothalamus may influence motor activity via the pineal gland and related epithalamus. (From Toni R. “Il sistema ipotalamo-ipofisario nell’antichità [The hypothalamic-pituitary system in the antiquity] - Dedicato alla memoria del Prof. Aldo Pinchera [Dedicated to the memory of Prof. Aldo Pinchera], In: L’Endocrinologo, Per una Storia dell’Endocrinologia [For a History of Endocrinology], 13, suppl. to n. 6, 1-11, 2012.

 

The current term “hypothalamus", however, was not actually introduced until 1893 by the Swiss anatomist, Wilhelm His.  On the basis of his studies on the ontogenesis of the human, fetal brain, His named the first anatomical subdivision of the hypothalamus the “pars optica hypothalami” (5), which is now recognized to include the preoptic region, tuber cinerium and infundibulum.  Discovery of the connection between the hypothalamus and posterior pituitary (supraoptic-hypophysial tract) by Ramon Cajal in 1894, and subsequent work on neurosecretion in fish hypothalamus by the Sharrers in 1928, set the groundwork for rapid advancement in the understanding of the hypothalamus that unraveled throughout the 20th century and continues into the 21st century.  Table 1 summarizes the major historical advances in the elucidation of the functional anatomy of the mammalian hypothalamic-pituitary unit (6).

 

Table 1. Timeline of Major Breakthroughs in Elucidation of the Functional Anatomy of the Mammalian Hypothalamic-Pituitary Unit

II century A.D.

Galen in Anatomicae Administrationes describes the third ventricle and its association with the rete mirabilis around the pituitary gland and dorsally with the pineal gland.  In De Usum Partium considers the hypothalamic infundibulum and pituitary gland as draining route and receptacle for brain mucous to the nasopharynx

1928

E. Scharrer describes “glandular cells” in the fish hypothalamus (concept of “neurosecretion”)

1316

Mondino de Liuzzi da Bologna in his Anothomia refers to the third cerebral ventricle as “integrator” of body functions, including psychic, emotional, and behavioral responses

1930

Popa and Fielding describe in the human pituitary stalk a portal vascular system interpreted as a route of the blood upward the hypothalamus

1522

Berangario da Carpi in his Isagogue Breves denies the existence of the Galenic  rete mirabilis in the human brain

1940-1955

Harris and Green establish the basis for the neural control of the pituitary gland secretion and demonstrate its vascular link with the hypothalamus

1543

Vesalius includes in the Fabricathe first anatomical drawings of the hypothalamic infundibulum, pituitary and their venous drainage

1954

WH Hess shows that both pituitary and autonomic responses are regulated by the anterior (trophotropic area) and posterior (ergotropic area) hypothalamus

1561-1627

Fallopius in the Observationes Anatomicae and Casserio in the Tabulae Anatomicaemention the arterial polygon at the base of the brain then described by Willis

1950-1958

Nauta and Kuypers describe the connections of the mammalian hypothalamus with the rest of the brain and propose that the limbic system influences pituitary function, introducing the concept of “hypothalamic integration”

1662

Descartes in his De Hominesuggests a connection between the optic nerve, third ventricle, and pineal gland to regulate body mouvments and coupling between neuroendocrine and motor responses in hypothalamic motivated behaviors

1960

Martinez describes the structure of the median eminence

1664

Willis in his Cerebri Anatomeargues that humors out of the third ventricle may be carried to the pituitary gland

 

1962

Halaz put forth the concept of “hypophysiotrophic area” of the hypothalamus

1655-1672

Schneider and Lower reject the Galenic idea that the pituitary gland filters brain secretions to the nose

1964

Szentagothi defines the tuberoinfundibular tract

1742

Lieutand discovers vessels in the pituitary stalk

 

 

 

1968

Guillemin and Schally isolate the first hypothalamic releasing factor

1767

Luigi Galvani in Disquisitiones Anatomicae circa Membranam Pituitariam discovers that mucus passing through the nostrils originates from small mucous glands of the human nasal mucosa and not from the pituitary

1969-1970

Yoshimura et al. show that mice pituitary chromophobes may behave like pituitary stem cells, and Nakane provides the first ultrastructural evidence for paracrine interactions in the pituitary gland

1778

Sommering introduces the term “hypophysis”

1971

L. Martini shows that hypothalamic releasing-factors regulate their own secretion via an “ultrashort feedback”

1787

 

 

 

Paolo Mascagni describes lymphatic vessels in human cranial  meninges, introducing the modern view of a lymphatic drainage of brain structures in mammals and man  

1984

T. Hokfelt demonstrates the presence of two different neurotransmitters in the same hypothalamic neuron, introducing the concept of “neuroendocrine regulation by multiple neuronal messengers”

1860

Von Luska describes the primary (or hypothalamic) capillary plexus of the portal vessels

1986

K. Fuxe and L. F. Agnati show that the median eminence is organized in modules, introducing the concept of “medianosome”, and hypothalamic neurons are regulated by both autocrine/paracrine and synaptic mechanisms, better known as “volume and wiring transmissions”

1872-1877

Meynert and Forel define the anatomical borders of what they call “the neural portion extending forward the region of the subthalamus” (i.e., the hypothalamus)

2009

Garcia-Lavandeira et al. identify stem cells/progenitors in the marginal zone of the adult human pituitary gland

1893

His introduces the term “hypothalamus” and provides the first anatomical subdivision based on ontogenesis of the human brain

2012

Iliff et al. describe the “glymphatic system” in rodents, glia-dependent perivascular tunnels interconnected with meningeal lymphatics, allowing for distribution of metabolites and neuromodulators to hypothalamic neurons

 

 

 

 

(Based on Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective.  J Endocrinol Invest 27, supp to n.6, 73-94, 2004, and Toni R. “Il sistema ipotalamo-ipofisario nell’antichità [The hypothalamic-pituitary system in the antiquity] - Dedicato alla memoria del Prof. Aldo Pinchera [Dedicated to the memory of Prof. Aldo Pinchera], In: L’Endocrinologo, Per una Storia dell’Endocrinologia [For a History of Endocrinology], 13, suppl. to n. 6, 1-11, 2012

 

ANATOMY OF THE PITUITARY GLAND

 

Gross and Radiologic Anatomy

 

The pituitary gland lies within a recess of the median part of the middle cranial fossa in the sphenoid bone (sella turcica) and is composed of two major components, the anterior lobe (adenohypophysis) and the posterior lobe (neurohypophysis) that can be readily distinguished radiologically by magnetic resonance imaging (Fig. 6).  The anterior lobe contains three subdivisions including the pars distalis, pars intermedia and pars tuberalis.  The pars distalis makes up the bulk of the anterior pituitary and is responsible for the secretion of anterior pituitary hormones into the peripheral circulation.  In man, it contains follicles of different sizes, typically surrounded by folliculostellate (FS) cells (7).  The pars intermedia lies between the pars distalis and the posterior pituitary, representing what remains of the original Rathke’s pouch cleft (see section on Embyryologic Anatomy).  Although considered vestigial in man, it contains follicles enriched with FS cells, mainly at its perimeter (i.e., the marginal zone), likely functioning as a subpopulation of pituitary stem cells (8). The pars tuberalis is well defined in most mammalian species, including man, and surrounds the infundibular stem (9).  The floor of the sella, or lamina dura, abuts the sphenoid sinus, allowing direct surgical access to the pituitary by the transsphenoidal route.  Other important boundaries to the pituitary gland are the cavernous sinus laterally, which contain the internal carotid artery surrounded with sympathetic fibers, and the cranial nerves III, IV, V (ophthalmic and maxillary branches), and VI (Fig. 7).  The optic chiasm is located superiorly, separated from the pituitary by the cerebrospinal fluid-filled suprasellar cistern and the dural roof of the pituitary, the diaphragma sella.

 

Figure 6. (A) Magnetic resonance image (MRI) and (B) corresponding schematic illustration of the human hypothalamus (H) and pituitary gland seen in sagittal orientation. Note the high intensity or "bright spot" of the posterior pituitary by MRI in (A), sharply defining the boundary between the anterior pituitary gland. III = third ventricle (Modified from Lechan RM. Neuroendocrinology of Pituitary Hormone Regulation. Endocrinology and Metabolism Clinics 16:475-501, 1987.)

Figure 7. (A) MRI and (B) schematic image of the pituitary fossa and its anatomic relationships seen in coronal orientation. The cavernous sinus contains the internal carotid artery and cranial nerves III, IV, V1, V2, and VI. The optic chiasm resides immediately above the pituitary gland and is separated from it by a cerebrospinal fluid-filled cistern. (Modified from Lechan RM. Neuroendocrinology of Pituitary Hormone Regulation. Endocrinology and Metabolism Clinics 16:475-501, 1987.)

Embryologic Anatomy

 

The posterior lobe of the pituitary gland is smaller than the anterior lobe and embryologically derives from the neural primordia as an outpouching from the floor of the third ventricle.  As a direct, anatomic extension of the central nervous system, it is not surprising that the posterior pituitary is composed primarily of unmyelinated axons and axon terminals as well as specialized glial cells called pituicytes.

 

In contrast to the posterior pituitary, the anterior pituitary derives from the oral ectoderm as Rathke's pouch, first seen by the third week of pregnancy in man, and gives rise to both the pars distalis and tuberalis.  There is little if any direct nervous innervation to the pars distalis, but cell to cell contact with the neuroectoderm of the primordium of the ventral hypothalamus is critical for differentiation of the anterior pituitary into the five major cell types.  This occurs as a result of the release of specific growth and transcription factors such as bone morphogenic protein (BMP)-4 and fibroblast growth factor (FGF)-8 (10).  Among the numerous transcription factors involved in positional determination and terminal differentiation of pituitary cell types (Fig. 8), the Notch signaling pathway serves a major role in mediating epigenetic regulation of lineage commitment through activation of non-coding RNAs and chromatin-histone interactions (11,12).  Recent evidence has also indicated a key role for SOX 2 and SOX3 in regulating pituitary morphogenesis both in rodent and man (13).  In humans, mutations of early transcription factors like Rpx, Prop-1 and Pit-1 lead to variable degrees of pituitary insufficiency (10).  Once the pituitary matures, the ability of the hypothalamus to communicate with the pars distalis is dependent upon the hypophysial portal system, a vascular link that connects the base of the hypothalamus to the pituitary gland.

 

Figure 8. Signaling molecules and transcription factors involved in the development of the mouse anterior pituitary from Rathke’s pouch. In the anterior lobe somatotrophs, lactotrophs and caudally-placed thyrotrophs derive from a common lineage, determined by Prop-1 and Pit-1. Independent lineages are observed for a rostrally-placed group of thyrotrophs, corticotrophs, gonadotrophs and intermediate lobe melanotrophs. All cell types are committed to a specific lineage through activation of Notch signaling at the placodal stage. (Adapted from Cohen and Radovick, Endocrine Reviews 23: 431-442, 2002; Zhu X, Gleiberman AS, Rosenfeld MG, Physiol Rev 87: 933-963, 2007; Zhu X, Wang J, Ju B-G, Rosenfeld MG, Curr Op Cell Biol 19: 605-611, 2007).

 

Microscopic Anatomy

 

Microscopically, the anterior pituitary is composed of nests or cords of cuboidal cells organized near venous sinusoids lined with a fenestrated epithelium into which secretory products from the anterior pituitary are collected.  Classically, five cell types and six secretory products of the anterior pituitary gland can be identified immunocytochemically including the somatotrophs (growth hormone), lactotrophs (prolactin), corticotrophs (adrenocorticotropic hormone), thyrotropes (thyroid-stimulating hormone), and gonadotrophs (luteinizing hormone and follicle-stimulating hormone) (14).  It is well recognized, however, that the anterior pituitary is vastly more complicated.  In addition to morphological and physiological evidence for heterogeneity among the classical anterior pituitary cell types (15-18) and the presence of clusters of a unique cell type, the folliculo-stellate cell (19), the anterior pituitary can also synthesize numerous other nonclassical peptides, growth factors, cytokines, binding proteins and neurotransmitters listed in Table 2 that are important for paracrine and/or autocrine control of anterior pituitary secretion and/or cell proliferation under defined physiological conditions (20).  Pituitary stem cells have now been recognized in adult mammalian pituitaries as a group of Notch-, Shh-, Wnt- and Hes1-positive elements without hormonal production, primarily residing in the marginal zone around the pituitary cleft (21).  However, it is possible that more than a single stem cell type is present in the anterior pituitary.   In fact, in rodents, a number of cell groups with stemness potential have been identified, including a subpopulation of folliculostellate cells having the ability to form cell colonies in vitro, a heterogeneous SOX2-positive, SOX9-negative, sphere-forming cell population, a Nestin-positive, potentially adult, progenitor group, and GFRa2-positive (Glial cell line-derived neurotrophic Factor Receptor), sphere-forming cells with clear features of multipotent elements (22).  GFRa2-positive cells have also been observed in the marginal zone of the adult, human pituitary (23).

 

Table 2. Nonclassical Anterior Pituitary Substances and Cell(s) of Origin

Substances

Cell Types

PEPTIDES

ACTIVIN B, INHIBIN, FOLLISTATIN

F, G

ALDOSTERONE STIMULATING FACTOR

UN

ANGIOTENSIN II (ANGIOTENSINOGEN, ANGIOTENSIN I

--

CONVERTING ENZYME, CATHEPSIN B, RENIN)

C, G, L, S

ATRIAL NATURETIC PEPTIDE

G

CORTICOTROPIN-RELEASING HORMONE-BINDING PROTEIN

C

DYNORPHIN

G

GALANIN

L, S, T

GAWK (CHROMOGRANIN B)

G

GROWTH HORMONE RELEASING HORMONE

UN

HISTIDYL PROLINE DIKETOPIPERAZINE

UN

MOTILIN

S

NEUROMEDIN B

T

NEUROMEDIN U

C

NEUROPEPTIDE Y

T

NEUROTENSIN

UN

PROTEIN 7B2

G, T

SOMATOSTATIN 28

UN

SUBSTANCE P (SUBSTANCE K)

G, L, T

THYROTROPIN RELEASING HORMONE

G, L, S, T

VASOACTIVE INTESTINAL POLTPEPTIDE

G, L, T

GROWTH FACTORS

BASIC FIBROBLAST GROWTH FACTOR

C, F

CHONDROCYTE GROWTH FACTOR

UN

EPIDERMAL GROWTH FACTOR

G, T

INSULIN-LIKE GROWTH FACTOR I

S, F

NERVE GROWTH FACTOR

UN

PITUITARY CYTOTROPIC FACTOR

UN

TRANSFORMING GROWTH FACTOR ALPHA

L, S, G

VASCULAR ENDOTHELIAL GROWTH FACTOR

F

CYTOKINES

INTERLEUKIN-1 BETA

T

INTERLEUKIN-6

F

LEUKEMIA INHIBITORY FACTOR

C, F

NEUROTRANSMITTERS

ACETYLCHOLINE

C, L

NITRIC OXIDE

F

C = corticotroph, F = folliculostellate cell, G = gonadotroph, L = lactotroph,

S = somatotroph, T = thyrotroph, UN = unknown

 

Blood Supply

 

The pars distalis of the anterior pituitary gland receives little or no arterial blood supply from branches of the internal carotid artery (24,25), while the posterior pituitary is fed by an anastomotic arterial circle derived from each of the inferior hypophysial arteries as they pierce the cavernous sinus (Fig. 9).  Rather, the pars distalis is supplied by venous blood delivered through the long portal veins that descend along the ventral surface of the pituitary stalk and interconnect capillary beds in the pars distalis with specialized capillary beds of the portal capillary system in the base of the hypothalamus called the median eminence (Fig. 9).  In turn, the portal capillary plexus in the median eminence receives arterial blood from a separate branch of the internal carotid artery, the superior hypophysial artery, after the internal carotid artery ascends from the cavernous sinus.  In addition to venous blood draining from the hypothalamus, the pars distalis also receives venous blood draining from the posterior pituitary through the short portal vessels, giving rise to approximately 30 per cent of the total blood supply to the anterior pituitary (26,27).  The perfusion sequence of arterial blood first reaching the posterior pituitary and the median eminence, followed by venous drainage to the anterior pituitary can visualized in man using rapidly enhanced magnetic resonance images (dynamic MRI) (28) (Fig. 10).  As a result of the venous blood flow pattern to the pituitary, the pars distalis is in a unique position where it can receive humeral information from the hypothalamus and the posterior pituitary, as well as substances circulating in the peripheral bloodstream.  Due to the location of pars tuberalis cells in the pituitary stalk and ventral surface of the median eminence, adjacent to the portal capillary plexus, it is likely that these cells also contribute to the humeral substances that are carried by a vascular route to the pars distalis (29), although its physiological significance is unknown.

Figure 9. Drawing of the vasculature of the primate anterior and posterior pituitary gland. A portion of the pituitary stalk (I) has been cut away to visualize the infundibular recess (IR) and portal capillaries (PC). CPV = confluent pituitary veins, CS = cavernous sinus, H = hypothalamus, IC = internal carotid artery, IHA = inferior hypophysial artery, IP = infundibular processes or posterior pituitary, LPV = long portal veins, SHA = superior hypophysial artery, SPV = short portal veins. (From Lechan RM, Functional Microanatomy of the Hypophysial-Pituitary Axis, in Melmed, S (Ed), Oncogenesis and Molecular Biology of Pituitary Tumors, Frontiers of Hormone Research, 20: 2-40, 1996.)

Figure 10. (A-D) MRI of sequential sequences of the stalk and pituitary gland in sagittal orientation following the intravenous administration of gadolinium. (A) Appearance prior to gadolinium. (B) Following gadolinium, the posterior pituitary is the first structure to show contrast enhancement. (C) This is followed by the pituitary stalk (arrow) and then finally (D) the anterior pituitary. (From Yuh et al, AJNR 15: 101-108, 1994.)

Venous drainage from the anterior pituitary to the systemic circulation is through adenohypophysial veins located at a sulcus separating the anterior pituitary from the posterior pituitary (24).  Other than the short portal vessels, venous drainage from the posterior pituitary collects into neurohypophysial veins, which together with adenohypophysial veins, extend as common vessels (confluent pituitary veins) to the cavernous sinus (Fig. 9).  The cavernous sinus is enriched by an additional draining system composed of paravascular spaces around the hypophysial arteries and veins, variably interconnected with intradural channels lined by a lymphatic endothelium, giving rise to the pituitary glymphatic system (30).  It provides a route for transport and clearance of intercellular liquids and metabolites into the blood circulation, allowing for volume transmission of soluble information at the level of the hypothalamic-pituitary unit (for anatomical details of this circuitry, see the section on the Glymphatic system and sinus-associated dural glymphatics).

 

ANATOMY OF THE HYPOTHALAMUS

 

Gross Anatomy

 

The hypothalamus lies directly above the pituitary gland (Fig. 11) and occupies approximately 2 per cent of the brain volume.  It is composed of a number of cell groups (Fig. 12) as well as fiber tracts that are symmetric about the third ventricle.  In sagittal section, the hypothalamus extends from the optic chiasm, lamina terminalis and anterior commissure rostrally to the cerebral peduncle and interpeduncular fossa caudally (Fig. 11).  The cavity of the third ventricle lies in the midline.  In coronal section (Fig. 13), each of the two symmetric walls of the hypothalamus can be divided into four surfaces: a lateral surface contiguous with the thalamus, subthalamus and internal capsule, the latter dividing the hypothalamus from the corpus striatum; a medial surface extending to the wall of the third ventricle, covered by ependymal cells; a superior surface corresponding to the hypothalamic sulcus that separates the hypothalamus from the central mass of the thalamus; and an inferior surface that is in continuity with the floor of the third ventricle.  The external surface of the hypothalamic floor (Fig. 14) gives rise to a median protuberance called the tuber cinereum (or gray swelling due to the pale bluish color of the blood vessels seen in the postmortem human brain), whose central part extends anteriorly and downward into a funnel-like process, the infundibulum or median eminence.  The infundibulum is in direct continuity with the infundibular stem of the posterior pituitary gland, and together with the pars tuberalis of the anterior pituitary, forms the pituitary stalk (Fig. 6).  Two additional symmetric eminences, the lateral eminences, corresponding to the most lateral portion of the hypothalamic wall and the postinfundibular eminence, as well as the symmetric mammillary bodies, complete the macroscopic morphology of the hypothalamic floor.

Figure 11. Midsagittal section of the human brain (from the XIX century wax collection of human brains at the Museum of the Department of Human Anatomy of the University of Bologna, Italy). The hypothalamus (asterisk) lies above the pituitary gland (cross) and has as its boundaries (1) the anterior commissure and lamina terminalis anteriorly; (2) mammillary bodies and midbrain posteriorly, and (3) thalamus superiorly. (From Lechan R.M. and Toni R., Regulation of Pituitary Function, in Korenman S.G (Ed), Atlas of Clinical Endocrinology, Current Medicine, vol IV, 1-25, 2000).

Figure 12. Magnified view of a fixed human brain in midsagittal orientation. The third ventricle makes up the core of the hypothalamus and extends into the pituitary (or infundibular) stalk, creating the infundibular recess. Many of the major cell groups are located near the midline. These include (from rostral to caudal) the preoptic nucleus (Pop), paraventricular nucleus (Pvn), dorsomedial nucleus (Dm), ventromedial nucleus (Vm), arcuate (or infundibular) nucleus (If), posterior hypothalamic nucleus (Po), and medial mammillary nucleus (mm). Ac = anterior commissure, fx = fornix, lt= lamina terminalis, ot = optic tract and chiasm, Lv = lateral ventricle, MB = midbrain, PN = pons, Sr = supraoptic recess, T = thalamus. (From Lechan R.M. and Toni R., Regulation of Pituitary Function, in Korenman S.G (Ed), Atlas of Clinical Endocrinology, Current Medicine, vol IV, 1-25, 2000).

Figure 13. Coronal section of a fixed human brain at the level of the posterior hypothalamus. The third ventricle (III) lies in the midline directly above the mammillary bodies (m). The subthalamus (sb), zona incerta (zi) and thalamus (T) are located at the superior border of the hypothalamus, whereas the corpus striatum (ST) is located laterally. FL = fasciculus lenticularis, FT = fasciculus thalamicus, ic = internal capsule, SN = substantia nigra, H1 = field H1 of Forel; H2 = field H2 of Forel. (From Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.)

Figure 14. Basal view of the brain showing the external surface of the floor of the hypothalamus and its arterial vessels. The infundibulum (I) lies posteriorly to the optic tracts and chiasm (ot) and anterior to the mammillary bodies (m). The arterial circle of Willis surrounds the hypothalamic floor and provides the arterial supply to the hypothalamic nuclei and fiber tracts. ac = anterior cerebral artery, aco = anterior communicating artery, b = basilar artery, ic = internal carotid artery, P = pons, pc = posterior cerebral artery, pco = posterior communicating artery. (From the XIX century wax collection of human brains at the Museum of the Department of Human Anatomy of the University of Bologna, Italy.)

Embryologic Anatomy

 

The diencephalon derives from the caudal part the pro-encephalic vesicle, which is the cranial expansion of the primitive neural tube, and the hypothalamus develops from the lateral wall of the diencephalon by extending ventrally to a groove called the “hypothalamic sulcus” that appears early in the lateral wall of the diencephalon (Figure 15).  Therefore, the hypothalamus can be considered a ventral derivative of the neural tube and to originate from the embryonic basal plate (31).  Since the basal plate is the source of all skeletal and autonomic motor neurons in the CNS, by inference, the hypothalamus has also been considered a motor system (32).  Indeed, neuroendocrine neurons that are involved in the regulation of the anterior and posterior pituitary secretion clearly have secretomotor functions.  However, some authorities believe that the basal (motor) plate of the neural tube ends at the level of the mesencephalon, and that the diencephalon (hypothalamus included), is actually a derivative of the dorsal or alar plate, which is primarily sensory (33).  Partial confirmation of this idea has been recently provided by the evidence that mouse embryonic stem cells may spontaneously differentiate into neurons expressing the Rax gene, a marker common to both the preoptic / tuberal hypothalamus and neural retina (a sensory structure), but do not express the Irx3 and En2 genes, typical of the midbrain structures (34). These findings are consistent with the presence of neurosecretory cells with sensory properties in the forebrain of invertebrates and fish (35), suggesting evolutionarily conserved sensory properties of neuroendocrine hypothalamic cells.

Within the neural tube, dividing hypothalamic neuroblasts remain confined within the cell layer adjacent to the ependymal canal (ependymal or ventricular layer), whereas postmitotic elements migrate more laterally into a cell-dense region (mantle layer) before reaching their final destination (36) (Figure 16).  Collectively, hypothalamic progenitors reach their final patterning and location in ventral and dorsal regions by exposure to two, specific, transcription factors, the ‘ventralizing’ Shh and subsequently the ‘dorsalising’ Bmp7.  As a result, specific transcription factor codes are established, leading to early differentiation of ventrolateral progenitors expressing the homeobox gene product, Pax 7 (37).  Outgrowth of neural process occurs at the most lateral borders of the hypothalamic mantle layer to give rise to tangential fiber tracts that course parallel to the ependymal canal and connect hypothalamic neurons with cranial and caudal portions of the developing neural tube.  These fiber tracts are highly ordered into spatial and temporal patterns (38).  Early connections include those with the midbrain (mammilotegmental tract) and hippocampus (stria terminalis), followed by those with the thalamus (mammilothalamic tract) (39).

Figure 15. Three-dimensional reconstruction of the developing proencephalon in the human embryo. Note that at the level of the inferior portion of the lateral wall is the region of the hypothalamus (Hyp) with the infundibular bud (I) and pituitary anlage (P) (Redrawn from Hines M, J Comp Neurol 34: 73-171,1922.) ap = alar plate, bp = basal plate, ce = cerebral hemisphere, cp = choroidal plexus, CS = corpus striatum, ep = epiphysis, EP = epithalamus, eps = epithalamic sulcus, h = hippocampal fissure, hs = hypothalamic sulcus, if = interventricular foramen, lt = lamina terminalis, oc = optic chiasm, sl = sulcus limitans, sr = supraoptic recess, T = thalamus.

Figure 16. Coronal section of the anterior hypothalamus in a human fetus of gestational age 12-14 weeks, counterstained with methylgreen and thionine. (A) Note that from the wall of the third ventricle, constituting the ependymal layer of the neural tube, a front of developing cells (arrows) migrate laterally towards the mantle layer to give rise to the primordium of the paraventricular nucleus (PVN). (B) High magnification of the image included in the rectangle shown in A. Note the high cellular density in the ependymal layer (EL) of the neural tube contrasts with the more diffuse distribution of migrating neuroblasts in the developing mantle layer (ML). III = third ventricle.

Organization of the hypothalamus into specific nuclear groups occurs in a temporal and spatial pattern both in rodents (38,39) and man (40), such that the entire preoptic to posterior lateral hypothalamus followed by the medially-located, neurohypophysial centers and the main part of the medial preoptic and tuberal hypothalamus all arise during an early phase of development, whereas the periventricular hypothalamus, the floor of the third ventricle and mammillary complex develop later (see Section C, Microscopic Anatomy).  Peak birth dates of specific hypothalamic nuclei in the primate are shown in Table 3.

 

Table 3.  Birthdates of Hypothalamic Nuclei in the Primate Brain

Hypothalamic nucleus

Peak birthdate

MPA

e43-e45

SCN

e30-e43

SON

e30-e38

PVN

e40-e43

ARC

e30

VMN

e30

DMN

e38

(Based on van Eerdenburg FJCM, Rakic P.  Early neurogenesis in the anterior hypothalamus of the rhesus monkey.  Dev. Bran Res. 79: 290-296, 1994)

 

 In addition to generalizations above regarding the development of specific hypothalamic nuclei, there are developmental differences that distinguish neuroendocrine neurons in the hypothalamus from non-neuroendocrine neurons.  Namely, neuroendocrine neurons, including those that give rise to the tuberoinfundibular and magnocellular neurohypophysial systems that are involved in regulation of the anterior and posterior pituitary, respectively (see later), differentiate immediately after closure of the neural tube, even before reaching their final destination within hypothalamic nuclei (41).  This phenomenon has been clearly demonstrated for GnRH neurons, that are fully differentiated at the level of the olfactory placode, even before migrating into the preoptic region of the hypothalamus (42).  Similarly, neuroblasts immunoreactive for the hypophysiotropic peptides, somatostatin and thyrotropin-releasing hormone, can be identified in the human fetal hypothalamus at the interface between the ependymal and mantle layers during a developmental stage that precedes complete formation of the PVN (43,44).

 

A number of genes have now been identified that regulate the temporal and spatial patterns of differentiation of hypothalamic cell groups.  The POU III-related homeobox genes, Brn-1, Brn2, and Brn4, are involved in the development of the periventricular and medial parts of the hypothalamus (45).  Transgenic mice with loss of function mutations or with targeted disruption of the Brn-2 gene, lack both the PVN and supraoptic nuclei, and have no somatostatin-producing neurons in the periventricular hypothalamus (46,47).  Expression of Brn-2 is dependent upon transcription factors Sim1 and ARNT2, since mutations of these genes in transgenic mice result in a phenotype that is similar to the Brn-2 KO mice (48-50).  Similarly, mice with decreased Sim1 expression have reduced vasopressin and oxytocin neurons, and develop a hyperphagic and obese phenotype (51).  A number of other genes have been identified that are involved in differentiation of specific hypothalamic nuclei and are listed in Table 4.  Temporal and spatial expression of many of these genes is selectively regulated by circulating sex hormones (52) and peripheral satiety signals such as leptin (53), suggesting that innate neuroendocrine behavioral responses are epigenetically influenced during the embryonic and fetal life.  Indeed, epigenetic imprinting in the mammalian hypothalamus has been recognized for a number of maternally silenced genes by knockout of the paternally-expressed allele including: a) Magel2, encoding a transcriptional regulator whose disruption leads to neonatal growth retardation, excessive post-weaning weight gain, adiposity, reduced food intake, and disappearance of orexin neurons; b) Ndn, encoding the growth suppressor and anti-apoptotic factor, necdin, whose loss leads to reduction in oxytocin and LHRH neurons; c) Nnat, encoding neuronatin that regulates energy homeostasis for which a single nucleotide polymorphism in man is associated with severe childhood and adult obesity; d) Gnasxl, encoding the transcript of the Gsα isoform, XLαs, whose inactivation results in a hypermetabolic phenotype with decreased adiposity, increased glucose tolerance and insulin sensitivity; and e) Pag3, that encodes a Kruppel-type zinc finger transcription factor whose absence reduces metabolic rate, lowers the core body temperature, increases adiposity, induces leptin resistance, reduces sympathetic activity, and alters the proportion of neuropetide neurons in the periventricular and medial hypothalamic nuclei.  Paternally-imprinted genes have also been recognized in the hypothalamus including Gnas that has selective expression in the PVN and whose constitutive knockout leads to Albright hereditary osteodystrophy characterized by severe obesity, lethargy, glucose intolerance and insulin, TSH and PTH resistance (54).

 

Table 4. Genes and Transcription Factors Involved in the Development of Specific Regions of the Rodent Hypothalamus

Gene

Nuclear Region

Brn-1, Brn-2, Brn-4

PVN, SON, PV, POA, MN, PH

Dlx1

TH

Vgll2, SF-1, Sox14, Satb2,

VMN

Fezf1, Nkx2-2, COUP-TFII

 

Gsh1, Mash1

ARC, VMN

Otp

PVN, SON, PV, POA, AH, ARC

rPtx-2

TH, MN

Sim1

PVN, SON

Fkh5

MN

Tst-1

MN, PH

(Based on Markakis E. A. Frontiers in Neuroendocrinology, 23: 257-291, 2002; McNay DE, Pelling M, Claxton S, Guillemont F, Ang S-L, Mol Endocr 20: 1623-1632, 2006; Kurrasch DM, Cheung CC, Lee FY, Tran PV, Hata K, Ingraham HA. J Neurosci 27: 13624-13634, 2007.)

AH = anterior hypothalamus, ARC = arcuate nucleus, MN = mammillary nuclei = posterior hypothalamus, POA = preoptic area, PV = periventricular nucleus, PVN = paraventricular nucleus, SON = supraoptic nuclei, TH = tuberal hypothalamus, VMN = ventromedial nucleus

 

Microscopic Anatomy

 

BOUNDARIES AND ORGANIZATION OF NEURONAL CELL GROUPS

 

Using phylogenetic and cytoarchitectonic criteria (55), a number of nuclear groups and fiber tracts are recognized in the vertebrate hypothalamus.  These are organized into three major regions including the lateral, medial and periventricular hypothalamus, each having distinct morphological and functional features.  In the human hypothalamus, the anterior column of the fornix that extends rostro-caudally through the substance of the hypothalamus to end in the mammillary bodies, and the mammillo-thalamic tract that projects from the mammillary bodies upward to the thalamus, create an anatomical boundary that divides the hypothalamus into medial and lateral subdivisions (Fig. 17).  Contained within the medial subdivision is the periventricular subdivision, a 5-6 cell layer thick nuclear group surrounding the third ventricle that is easily recognized in rodents using standard vital stains, but has less clear anatomical boundaries in the human brain.

 

Both the medial and periventricular subdivisions of the mammalian hypothalamus contain a high density of neuronal cell bodies organized into nuclear groups (Tables 5 and Fig. 17) and in the human brain, has been classified with a number of different synonyms (Table 6).  Both subdivisions are crucial for the regulation of the anterior and posterior pituitary gland.  The medial hypothalamus also contains nuclear groups that serve as relay centers for highly differentiated neural information coming from the neocortex, limbic system and autonomic sensory centers in the brainstem involved in initiation phases of specific homeostatic behaviors such as thirst, hunger, thermoregulation, the sleep-wake cycle, and reproductive behavior (55). The lateral hypothalamus occupies the largest portion of the hypothalamus by volume.  However, it has relatively fewer neurons compared to the medial hypothalamus, and only a limited number of nuclear groups intercalated within a massive fiber system, the medial forebrain bundle (MFB).  It is through this fiber system that information from the medial forebrain (amygdala, hippocampus, septum, olfactory system, neocortex) and the brainstem is carried to the medial and periventricular hypothalamic subdivisions, delegating an important role to the lateral hypothalamus to influence homeostatic control systems elaborated by the medial hypothalamus.  Figure 18 schematically depicts major interrelationships between the periventricular, medial and lateral hypothalamic subdivisions and the rest of the brain.

Figure 17. Schematic representation of the human hypothalamus in coronal orientation (A-D: rostral to caudal), demonstrating the location of major nuclear groups. Drawings correspond to MRI images in Fig. 26. Using the fornix (fx) as an anatomic landmark as it passes through the mid-portion of the hypothalamus on each side of the third ventricle, it is convenient to divide the hypothalamus into medial and periventricular zones (that lie largely medial to the fornix) and a lateral zone (that lies lateral to the fornix). The medial and periventricular zones contain most of the hypothalamic cell groups, and the lateral zone contains relatively fewer neurons. This is because the lateral zone is largely composed of a massive bidirectional fiber pathway – the medial forebrain bundle – that extends through the hypothalamus and interconnects it with the limbic system and brainstem autonomic centers.

 

Table 5. Major Hypothalamic Cell Groups in Mammals

PERIVENTRICULAR ZONE

PERIVENTRICULAR NUCLEUS

SUPRACHIASMATIC NUCLEUS

PARAVENTRICULAR NUCLEUS

ARCUATE NUCLEUS

MEDIAL ZONE

MEDIAL PREOPTIC NUCLEUS

ANTERIOR HYPOTHALAMIC NUCLEUS

DORSOMEDIAL NUCLEUS

VENTROMEDIAL NUCLEUS

PREMAMMILLARY NUCLEUS

MAMMILLARY NUCLEUS

POSTERIOR HYPOTHALAMIC NUCLEUS

LATERAL ZONE

LATERAL PREOPTIC NUCLEUS

LATERAL HYPOTHALAMIC NUCLEUS

SUPRAOPTIC NUCLEUS

rostral to caudal order of appearance in each zone

Based on the anatomical classification of Nauta WJH and Haymaker W, Hypothalamic nuclei and fiber connections.: Haymaker W, Anderson E, Nauta WJH (eds); The Hypothalamus, Charles C Thomas Publisher, 1969, pp 136-209

 

Table 6. Terminology of hypothalamic nuclei in the human brain (rostral to caudal order of appearance)

Spiegel
Zweig
1919

Clark
1936

Brockhaus
1942

Khulenbeck
Heimaker / Nauta
1949-69

Feremutsch
1955

Diepen
1962

Schattelbrand
Wahren
1977

Braak
1987

Swaab
1985-92

GTD

POA

n. prothal. periventr.

nn. lineae medianae
preoptic periventric.n.

a. periventr. hypothal.
e / ba

preoptic
groups

n. prothal. periventr.
d / int / v

periventricular n.

 

SCN

POA

n.ovoideus

SCN

a. periventr. hypothal.
communis

SCN

n. ovoideus

SCN

SCN

GTD

POA

n. prothal.
princip.
o / ce / v

medial preopt.ic n.
anterior hypothal. n.
periventr. preoptic n.
lateral preoptic n.

a. periventr. hypothal anterior
a. lateralis hypothal anterior
a. lateralis hypothal. reticularis

lateral
hypothal.
n.

n. prothal.
principalis
o / ce / v / l

chiasmatic grey
cuneiform n.
uncinate n.

POA
OVLT

GTD

POA

nucleus
intermed.

medial preopt. n.
anterior hypoth. n.

intermediate lateral hypothal. a.

 

n. prothal.
princip.
ce / v

intemediate n.

SDN

GTD

POA

orolateral
hypothal. n.

n. supraoptic
diffusum

     

retrochiasmatic
n.

 

SON

SON

SON

SON

SON

SON

SON

SON*

SON

PVN

PVN

PVN

PVN

PVN

PVN

PVN

PVN*

PVN

MII

   

arcuate
or infundibular n.

a. periventr. basalis posterior

INF

INF

INF°

 

GTD

VMN

 

VMN

a. lateralis hypothal. ventromed.

VMN

VMN

VMN, postero-medial n.

 

GTD

DMN

 

DMN

a. periventr. hypothal. communis
a. lateralis hypothal. posterior

DMN

DMN

DMN

 

GTD

LHA

 

TMN

mammillo-infundibular n.

TMN

TMN

TMN

 
 

PN

 

PN

     

PN

 

PFN

   

PFN

a. lateralis hypothal. posterior (parafornicalis)

PFN

PFN

   

GTD

   

DN

n. paraventricularis pars caudalis
a. lateralis hypothal. posterior
pars dorsalis

a. dorsalis

n. dorsalis

   

GTD

LHA

 

LHA

a. lateralis hypothal. reticularis
pars principalis

pars lateralis tubero-mammillaris

n. lateralis

   

nn. tuberis

nn
tuberis

 

nn. tuberis laterales

n tuberis lateralis hypothalami

nn. tuberis
lateralis

n. tuberis
lateralis

LTN

LTN

 

MMN

 

MMN

n. corporis mammillaris

 

MMN

   
 

LMN

 

LMN

   

LMN

   
 

n. interc.

 

n. interc.

n. interc.hypothal.

 

n. interc.

   

DMN dorsomedial nucleus, GTD = griseum tuberis diffusum, INF = infundibular nucleus, LHA = lateral hypothalamic area, LMN = lateral mammillary nucleus, LTN = lateral tuberal nucleus, MII = massa infundibularis intermedia, MMN = medial mammillary nucleus, OVLT = organum vasculosum lamina terminalis, PFN = perifornical nucleus, PN = posterior nucleus, POA = preoptic area, PVN = paraventricular nucleus, SCN = suprachiasmatic nucleus, SDN = sexual dimorphic nucleus. SON = supraoptic nucleus, TMN = tuberomammillary nucleus, VMN = ventromedial nucleus, a. = anterior; ce = centralis, d = dorsalis, int. = intermedius, l = lateralis, n. interc= nucleus intercalatus, nucleus intermed. = nucleus intermedius, n. prothal. periventr. = nucleus prothalamicus periventricularis, n. prothal. princip. = nucleus prothalamicus principalis, o = oralis, v = ventralis; * = associated with surrounding accessory magnocellular neurosecretory nuclei; ° = including cranially the periventricular nucleus. Based on Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27, supp to n.6, 73-94, 2004

Figure 18. Schematic representation of the major neural pathways connecting the periventricular, medial and lateral hypothalamic subdivisions with the rest of the brain. Groups with identical colors are functionally linked.

Each of the three hypothalamic subdivisions can be further divided along the rostral-caudal axis into the: a) anterior or chiasmatic region, extending between the lamina terminalis and the anterior limit of the infundibular recess; b) median or tuberal region, extending between the infudibular recess and the surface of the anterior column of the fornix; and c) posterior or mammillary region, extending between the anterior column of the fornix and the caudal limit of the mammillary bodies.

 

Recent tract-tracing and morphofunctional studies in rodents have proposed a functional perspective of hypothalamic subdivisions aimed at coordinating behavioral responses like feeding, reproduction and defense/exploration with autonomic and neuroendocrine responses.  In particular, it has been suggested that specific nuclei in the rostral part (chiasmatic region) of the periventricular subdivision (namely the preoptic area) and dorsal zone of the tuberal region (namely the dorsomedial nucleus) reciprocally interact to provide outputs unique for different pools of neuroendocrine neurons located along the walls of the third ventricle (collectively considered as a periventricular motor zone), coupled to outputs to selective pools of autonomic neurons in all hypothalamic subdivisions (collectively considered as preautonomic cell groups).  This neuronal network would be responsible for constant, reproducible but different patterns of endocrine and autonomic activation in response to specific homeostatic signals (hunger, sexual desire, motivated motor activity), constituting a hypothalamic visceromotor pattern generator (HVPG).  In this manner, it would be clearly recognized that the HVPG is comprised of a contingent of neurons interposed between the classical periventricular and medial hypothalamic subdivisions (56).

 

CIRUMVENTRICULAR ORGANS 

 

Median Eminence (ME)

 

One of the most important regions in the hypothalamus that is essential for regulation of the pituitary gland is the median eminence, a midline structure located in the basal hypothalamus ventral to the third ventricle and adjacent to the arcuate nucleus.  It is here that all hypophysiotropic hormones converge before they are conveyed to the pituitary gland.  The median eminence is one of seven so called circumventricular organs situated as midline structures in the walls of the lateral, third or fourth ventricles (57,58).  Other circumventricular organs include the organum vasculosum of the lamina terminalis, subfornical organ, choroid plexus, pineal gland, subcommissural organ and area postrema(Fig. 19).  Characteristically the circumventricular organs contain a rich capillary plexus and with the exception of the subcommissural organ, have a fenestrated endothelium rendering the structures outside of the blood brain barrier. This morphologic feature together with the presence of neural elements contacting the fenestrated capillaries allows the secretion of brain-derived products into the peripheral circulation and/or makes circumventricular organs targets for blood-born information which can then be transmitted to the brain (59).

Figure 19. Location of circumventricular organs in the rat brain. AP = area postrema, ME = median eminence, OVLT = organum vasculosum of the lamina terminalis, P = pineal gland, PP = posterior pituitary, SFO = subfornical organ. (Modified from Saper and Breder, New England Journal of Medicine 330: 1080-1886, 1994.)

The median eminence is a highly organized structure containing three zones: the ependymal zone, the internal zone (or zona interna) and the external zone (or zona externa) (60,61) (Fig. 20A).  The ependymal zone forms the floor of the third ventricle and has some very specialized features including densely formed tight junctions between adjacent cells and highly specialized cells, tanycytes, that extend bleb-like protrusions and microvilli into the cerebrospinal fluid (CSF) at their ventricular surface and long cytoplasmic processes ventrally into the substance of the median eminence (61,62).  Since the portal capillaries in the median eminence lie outside of the blood brain barrier, one of the functions of the ependymal zone is to create a barrier to the brain, preventing substances released into the periportal capillary spaces from entering the cerebrospinal fluid (63,64).  Tight junctions also can be found at the dorso-lateral margins of the median eminence adjacent to the neuropil of the arcuate nucleus created by perivascular tanycyte processes (48), thereby compartmentalizing all substances entering the periportal capillary spaces within the confines of the median eminence, itself.  Norsted et al (49), however, have demonstrated the absence of endothelial barrier antigen in blood vessels at the far ventro-medial aspect of the arcuate nucleus close to the border between the walls and floor of the third ventricle, and propose the lack of a blood brain barrier in this region.  Allowing blood-born substances including gut peptides, glucose, amino acids and fatty acids to enter the arcuate nucleus in this region may be an important homeostatic mechanism that contributes to the regulation of appetite and satiety (see later).

 

Figure 20. (A) Schematic diagram of the median eminence showing the organization of its three major zones: ependymal zone (E), internal zone (ZI), and external zone (ZE). ZE is invigilated by portal capillaries which are contacted by axon terminals of the tuberoinfundibular system and by processes of specialized ependymal cells, the tanycytes. (B) Fibers coursing through the ZI are seen immunocytochemically in the rat using antiserum to vasopressin. (C) Fibers terminating in the ZE in close association to portal capillaries (PC) are seen immunocytochemically in the rat using a proTRH-directed antiserum. III = third ventricle. (From Lechan RM, Functional Microanatomy of the Hypophysial-Pituitary Axis, in Melmed, S (Ed), Oncogenesis and Molecular Biology of Pituitary Tumors, Frontiers of Hormone Research, 20: 2-40, 1996.)

Based on their location, morphology, cytochemistry and ultrastructure, tanycytes can be divided into several subtypes including alpha, beta and recently characterized gamma subtypes (65,66).  The alpha subtypes line the ventrolateral walls of the third ventricle and beta subtypes line the floor and lateral extensions of the third ventricle.  Gamma subtypes are small tanycytes that can be found throughout the substance of the median eminence (67).  While presumed to be barrier cells, they likely have other important, neuroendocrine functions that may supersede their role as barrier cells.  The close association of tanycyte foot processes with the basal lamina of the portal capillaries and with individual axon terminals (Fig. 21) could create a retractable barrier to regulate the diffusion of secretory products entering or exiting specific regions of the portal capillary plexus or from axon terminals (68,69).  This mechanism has been shown to have an important role in the regulation of gonadal function in photoperiod sensitive animals, in which retraction of the tanycyte foot processes from portal vessels during long days allow activation of reproductive function (70).  A similar dynamic interaction between glial cells and secretory nerve endings in the posterior pituitary have been described by Beagley and Hatton (71).  In addition, the absorption of substances from the CSF at its apical surface for transport to the portal capillaries (62,72,73) could result in a mechanism whereby secretory products released into the CSF have access to the anterior pituitary.  Tanycytes may also serve as a scaffolding for axons entering the median eminence during embryologic development, guiding them to their ultimate destination in the external zone (74).  Tanycytes express one of the highest concentrations in the brain of type 2 deiodinase (D2) (75), the enzyme responsible for the conversion of thyroxine (T4) into its more biologically potent product, triiodothyronine (T3), the D2 degrading and reactivating enzymes, WSB-1 and VUD-1 (76), thyroid hormone transport (78.).  These observations among others are in keeping with recent reports on the important role of tanycytes in control of the hypothalamic-pituitary-thyroid axis and regulating tissue levels of thyroid hormone in the hypothalamus (79-81).

Figure 21. Electron micrograph of the external zone of the median eminence showing the presence of axon terminals (a) and a tanycyte process (t) adjacent to a fenestrated capillary (C) of the portal plexus. One axon (closed arrowhead) has been engulfed by the tanycyte and another (open arrowhead) is separated from the portal capillary space by the tanycyte foot process. Note presence of dense core vesicles (arrows) as well as smaller secretory vesicles in several axon terminals. (From Lechan RM, Functional Microanatomy of the Hypophysial-Pituitary Axis, in Melmed, S (Ed), Oncogenesis and Molecular Biology of Pituitary Tumors, Frontiers of Hormone Research, 20: 2-40, 1996.)

Tanycytes also express a number of embryotic genes (82), suggesting that they may serve as stem cells.  Indeed, any damage to tanycytes is repaired by rapid regeneration of these cells to reline the third ventricle (83).  Along these lines, tanycytes have been observed to express POMC mRNA (84), raising the possibility that they may have the ability to differentiate into neurons and contribute to the neuronal population in the adjacent arcuate nucleus.  Other evidence for tanycyte differentiation into neurons has also been given (82,85).  Finally, tanycytes have properties of inflammatory cells and may be capable of producing cytokines and chemokines that contribute to the mechanism of hypothalamic inflammation associated with a high fat intake (personal observations).

 

The internal zone of the median eminence lies directly below the ependymal zone and is primarily composed of unmyelinated axons of passage of the hypothalamic-neurohypophysial system en route to the posterior pituitary (Fig. 20B).  Characteristic of these axons are dilatations or Herring bodies, in which collect large numbers of neurosecretory granules measuring 200 to 350 nm in diameter (Inset, Fig. 20B).  The internal zone also contains cytoplasmic processes of tanycytes and axons of passage of the hypothalamic tuberoinfundibular system as they descend into the external zone.

 

The external zone underlies the internal zone and in addition to the portal capillaries and cytoplasmic extensions of the tanycytes described above, it contains numerous fine calibers, unmyelinated axons and axon terminals of the hypothalamic tuberoinfundibular system (Fig. 20C).  Characteristic of these axon terminals are dense-core vesicles ranging from 50 to 130 nm in diameter (Fig. 21).  The close proximity of many of the axon terminals to the portal system suggests that these axons are capable of secreting the material stored in their vesicles into the pericapillary spaces and by percolating through the fenestrated endothelium of the portal capillaries, reach the anterior pituitary by way of the long portal vessels.  These substances, commonly referred to as hypothalamic releasing and inhibitory hormones on the basis of their ability to stimulate or inhibit anterior pituitary hormone secretion respectively, have been chemically identified and are listed in Table 7.

 

Table 7. Classic Hypothalamic Releasing and Inhibitory Substances

Substance

Acids

CORTICOTROPIN-RELEASING HORMONE (CRH)
SER - GLU - GLU - PRO - PRO - ILE - SER - LEU - ASP - LEU - THR - PHE - HIS - LEU - LEU-ARG - GLU - VAL - LEU - GLU - MET - ALA - ARG - ALA - GLU - GLN - LEU - ALA - GLN -GLN - ALA - HIS - SER - ASN - ARG - LYS - LEU - MET - GLU - ILE - ILENH2

41

DOPAMINE

1

GROWTH HORMONE-RELEASING HORMONE (GHRH)
TYR - ALA - ASP - ALA - ILE - PHE - THR - ASN - SER - TYR - ARG - LYS - VAL - LEU - GLY - GLU - LEU - SER - ALA - ARG - LYS - LEU - LEU - GLN - ASP - ILE - MET - SER - ARG - GLU - GLN - GLY - GLU - SER - ASN - GLN - GLU - ARG - GLY - ALA - ARG - ALA - ARG - LEUNH2

44

GONADOTROPIN-RELEASING HORMONE (GnRH)
pyroGLU - HIS - TRP - SER - TYR - GLY - LEU - ARG - PRO - GLYNH2 -

10

SOMATOSTATIN
ALA - GLY - CYS - LYS - ASN - PHE - PHE - TRP - LYS - THR - PHE - THR - SER - SER – CYS S _____________________S

14

THYROTROPIN-RELEASING HORMONE
pyroGLU - HIS - PRONH2

3

 

Many axon terminals, however, do not abut directly on portal capillaries or terminate at some distance from the portal capillary plexus, may be to serve a modulatory role on other axon terminals rather than secrete into the portal plexus and explain the large numbers of peptides in the median eminence that either have no certain, direct action on anterior pituitary cells or cannot be measured in the portal blood (86).  Axon terminals containing dopamine, for example, are located in close proximity to axon terminals containing GnRH (87) at the lateral margins of the external zone of the median eminence and can modulate the secretion of GnRH by presynaptic inhibition (88,89).  Galanin containing axon terminals have also been observed to overlap with GnRH terminals in the lateral portion of the median eminence (90) but stimulate GnRH release from median eminence fragments (91).  Although axo-axonal synapses are uncommon in the median eminence of most animal species studied using morphologic criteria (54), receptors for several different peptide hormones have been identified on axon terminals in the external zone suggesting that axo-axonal interactions can take place.  Given the slow circulation time of blood perusing the median eminence (92), synaptic specialization in the median eminence may be unnecessary.

 

Alternatively, axons terminating at a distance from the portal capillaries may be held in reserve and only secrete to the anterior pituitary under certain physiological conditions.  This phenomenon has been described for several peptides such as neuropeptide Y, whose concentration increases in portal capillary blood during an ovulatory surge to potentiate the action of GnRH on gonadotropin secretion (93,94).  Similarly, VIP/PHI, which shows a minimal immunocytochemical staining pattern in the median eminence in the basal state, increases during suckling to stimulate prolactin release (95) and vasopressin markedly accumulates in the external zone following adrenalectomy (96).  The anatomical correlate of these physiologic observations may be suggested by the work by King and Letourneau (97) on gonadotropin regulation in which GnRH-containing axon terminals in the median eminence can be found at different distances from the portal capillaries in intact animals’ vs gonadectomized animals.  This indicates the potential for a dynamic association between axon terminals of the tuberoinfundibular system and the portal capillaries under specific physiologic conditions.  Marked reorganization in the median eminence of several different peptide-containing axon terminals in the median eminence has also been observed following hypophysectomy (98).

 

A further complexity to the physiology of axon secretion in the external zone of the median eminence is the common occurrence of more than one peptide or transmitter coexisting in the same axon terminal.  For example, TRH and preproTRH 160-169 coexist in the same axon terminals in the median eminence (99) and together have important potentiating effects on anterior pituitary TSH secretion (100).  Galanin coexists with GHRH in the majority of GHRH-tuberoinfundibular neurons (101) and although does not stimulate growth hormone secretion by itself in dispersed anterior pituitary cells (102), when administered together with GHRH, it increases GH secretion over what can be achieved by GHRH alone (103).  Rather than arise as a biosynthetic product of the same precursor molecule as preproTRH 160-169 and TRH, galanin and GHRH are derived from two separate gene products, expanding the possible sources for peptides that potentiate anterior pituitary secretion (104).  The coexistence of substances in axon terminals may also help to coordinate the secretion of separate anterior pituitary hormones as has been proposed for VIP/PHI, neurotensin, and enkephalin in CRH-producing neurons (105) to coordinate the secretion of ACTH, GH and prolactin during stress (106).

 

In addition to axon terminals in the external zone of the median eminence, densely packed fibers that contain VIP and the nitric oxide-synthesizing enzyme, nitric oxide synthase (107) have been described on the ventral surface of the median eminence separated from the external layer (108).  These fibers surround portal vessels and innervate smooth muscle of precapillary arterioles that supply the portal capillary plexus of the median eminence.  Since both VIP and NO are potent vasodilators (109,110), these substances may play an important role in regulating the rate of blood flow to the median eminence and hence to the anterior pituitary, thereby exerting a separate level of control over anterior pituitary secretion.  As opposed to axon terminals in the external zone of the median eminence that derive from the hypothalamus (see below), axons involved in regulation of portal blood flow appear to arise from other regions such as the sphenopalatine ganglion (107,108).

 

Consistent with the concept that the median eminence lies outside of the blood-brain barrier, claudin-5 and ZO-1, markers for tight junctions, are absent from vessels in the external layer (63).

 

Organum Vasculosum of the Lamina Terminalis (OVLT)

 

The OVLT is located in the midline of the lamina terminalis as part of the anterior wall of the third ventricle (Fig. 19).  Its dorsal surface protrudes into the third ventricle cavity and its ventral surface is in direct contact with the prechiasmatic cistern.  Thus, OVLT cells are in a position to be bathed by soluble factors in the CSF in both ventricular and cisternal spaces.  In rodents, ultrastructural studies by Weindl et al (111) and Mitro and Palkovits (112) have described a variety of cell types in the OVLT, including specialized neurons, tanycytes, ciliated ependyma, and glial cells (113).  Some of these cells send long processes to the periventricular space, whereas others establish specialized junctions and synaptic contacts or project outside the OVLT (113-115).

 

As in the median eminence, the OVLT contains fenestrated capillaries.  They are derived from small branches of the preoptic artery that break up into a dense network of small vessels in the pia matter lining the external surface of the lamina terminals, and loop up towards the ventricular lumen (116).  These vessels circumscribe interstitial spaces filled with cellular processes and secretory nerve endings that contain a number of neurotransmitter substances including atrial naturetic peptide, vasopressin, somatostatin, and GnRH (117), suggesting that like the median eminence, the OVLT subserves a neuroendocrine function.  In contrast to the median eminence, however, blood from the OVLT does not drain into a portal plexus, but rather primarily to the medial preoptic region (118), suggesting a close functional interrelationship between the OVLT and this region of the hypothalamus.  In addition, neurons in the OVLT project to the preoptic nucleus, subfornical organ, arcuate nucleus, supraoptic nucleus, medial thalamus and parts of the limbic system, primarily the cingulate, temporal and insular cortices.  This anatomical organization, therefore, strategically places the OVLT in an ideal location to receive blood-born information and then transmit this information to specific regions of the brain.  Accordingly, the OVLT has been implicated in mediating the febrile effects of circulating cytokines (see Thermoregulation).  In addition, the OVLT is involved in osmoregulation and fluid balance through osmoreceptor cells that express the transient receptor potential vanilloid (TRPV) 1 gene (119), and respond to circulating levels of angiotensin II and relaxin (120,121).  Its osmoregulatory role has been recently demonstrated in human volunteers subjected to excessive sweating using functional neuroimaging and blood flow distribution.  These studies showed that the OVLT is co-activated with limbic regions well known to be involved in thirst consciousness after thalamic relay (cingulate and temporal cortex), whereas after water ingestion, prominent activation of the cortical satiety centers (insula) occurred (122).  The OVLT is also densely innervated by axon terminals containing GnRH, originating from perikarya in the septum and areas surrounding the OVLT that presumably contribute to the regulation of pituitary gonadotropin secretion (123), perhaps through connections between the OVLT and the median eminence (124).  In female rodents, these axons cross the OVLT en route to the median eminence to trigger pulsatile proestral release of pituitary gonadotropins (125), whereas in males, their gonadotropin-releasing hormone content is regulated by levels of circulating thyroid hormone (126,127).  Direct connections between the OVLT and the median eminence have also been described (124). 

 

Subfornical Organ (SFO)

 

The name of this circumventricular organ derives from its midline, anatomical location under the fornix (Fig. 18), at the point where the lamina terminalis joins the tela choroidea of the third ventricle (128).  Embryologically, the SFO arises from the same part of the neural tube as the OVLT, and accordingly, have a similar microarchitecture and share common functions (129).

 

The SFO can be divided into two regions: a peripheral shell or “perimeter” that is rich in nerve endings arising from neurons intrinsic to the SFO but poor in blood capillaries, and a more densely packed center or “core” crowded with neuronal and glial perikarya and containing a dense vascular network of fenestrated and unfenestrated capillaries.  In caudal portions of the SFO, capillaries are continuous with those of the choroid plexus (130).  It is presumed that the “core” of the SFO is the locus for major hormonal receptor fields and fiber terminals of its afferent neuronal innervation, particularly the median preoptic nucleus, whereas the “perimeter” is the site of exit for SFO axons projecting to specific target regions in the hypothalamus including the preoptic nucleus, OVLT, supraoptic nucleus, paraventricular nucleus and lateral hypothalamus (131).

 

The SFO has an important role in coordination of fluid balance with blood pressure and drinking behavior, especially during hemorrhage and hypovolemia (132).  The rich vasculature of the SFO allows circulating angiotensin II to stimulate intrinsic neurons (133) via angiotensin type 1 receptors (134).  Through direct projections to the paraventricular nucleus, supraoptic nucleus and accessory magnocellular cell groups of the hypothalamus (135), SFO neurons induce release of vasopressin from the posterior pituitary (136), activate paraventricular nucleus neurons that descend to sympathetic centers of the spinal cord that regulate vasoconstriction (137), and possibly favor the release of vasoactive peptides like VIP from the anterior pituitary and a number of other neural sites related to fluid and blood pressure balance (138).  Evidence for intrinsic production of angiotensin II in the SFO (139) and the antagonistic effects of galanin released from axon terminals that synapse on SFO neurons on angiotensin II-induced drinking behavior and vasopressin release (140), may also contribute to the regulation of fluid homeostasis.  The presence of other peptides in the SFO and/or their receptors including obestatin, somatostatin and thyrotropin-releasing hormone, has suggested that the SFO might play a role in coordinating the ingestive behavior of liquids with solid food and the sleep cycle (141-143).  This idea is also supported by the presence of leptin receptors in the SFO, and that their deletion abolishes the leptin-mediated increase in sympathetic outflow to the kidney (144).  In addition, due to its connections with the preoptic nucleus and OVLT, the SFO is also involved in the regulation of thirst and locomotor behavior for drinking (145).

 

FIBER SYSTEMS

 

The fiber systems that link the hypothalamus to the rest of the brain are numerous and intricate, reflecting the importance of the hypothalamus as an integrating center for the rest of the brain.  Due to the complexity of the fiber systems, however, it is impractical to individually describe each fiber pathway linking each nuclear group, particularly for the human hypothalamus in which nuclear boundaries and relative projections are less clear than in other mammals.  Readers are referred to extensive reviews on this topic (146,147).  We will describe only the major hypothalamic fiber systems with respect to their afferent and efferent connections to the periventricular, medial and lateral hypothalamic nuclear subdivisions.

 

Afferent Connections

 

Inputs to the mammalian hypothalamus arise primarily from the limbic system, brainstem reticular formation, thalamus, subthalamus, basal ganglia, retina and possibly the neocortex (Fig. 22).  Afferents from the limbic system include 3 main fiber groups, the medial forebrain bundle, the stria terminalis, and the fornix.  The medial forebrain bundle is located in the most lateral part of the hypothalamus and contains fibers originating from more than 50 nuclear groups in different regions of the brain including descending fibers from the olfactory and septal areas, and ascending fibers from the amygdaloid complex and substantia innominata, the latter forming the ventral amygdalofugal component of the ansa peduncularis.  The stria terminalis originates in the amygdaloid complex, and the fornix in the hippocampus, both entering the rostral-medial hypothalamus close to the ventricular surface, and then arching into the substance of the hypothalamus to terminate along the entire extent of the hypothalamus.

 

Figure 22. Overview of the major afferent pathways to the hypothalamus. (A) Schematic organization of medial forebrain bundle (MFB). Fibers afferent to the hypothalamus enter the lateral wall of the hypothalamus and are shown in different colors in relation to their anatomical source (amygdala, septal areas, olfactory areas, frontal neocortex). Reciprocal efferent connections from the hypothalamus to the same regions are shown by dotted black lines parallel to the colored lines. Pink fibers (and related reciprocal black dotted lines) indicate the amygdalofugal (and related amygdalopetal) components of the ansa peduncularis, entering the hypothalamus as a part of the medial forebrain bundle. The mammillary body and anterior column of the fornix are colored light blue and lie medial to the course of medial forebrain bundle. (B) Schematic organization of limbic afferents to the hypothalamus via the fornix (fx), stria terminalis (st), stria medullaris (sm), and olfactory tract. Axons enter the rostral portion of the hypothalamus before coursing throughout its entire extent. (C) Course of afferent fibers from the thalamus, subthalamus and zona incerta to the hypothalamus. Efferents from the hypothalamus coursing in the mammillo-subthalamic tract are also shown. On the right side of the image, a three-dimensional reconstruction shows the anatomical structures schematically depicted on the left side. aap = amygdalofugal and amygdalopetal components of the ansa peduncolaris; ac = anterior commissure; Ah = Ammon horn; al = ansa lenticularis; am = amygdala; ap = ansa peduncularis; ATn = anterior thalamic nucleus; cc = corpus callosum. CN = caudate nucleus; CS = corpus striatum; df = dentate fascia; fl = fasciculus lenticularis; fr = fasciculus retroflexus; ft = fasciculus thalamicus; fx = fornix; H1 = field H1 of Forel; H2 = field H2 of Forel; ha = habenula; hipp = hippocampus; HYP = hypothalamus; ic = internal capsule; iTp-ap = inferior thalamic peduncle of the ansa peduncularis; LTn = lateral thalamic nucleus; mb = mammillary body; MFB = medial forebrain bundle; mst = mammillo-subthalamic tract; MTn = medial thalamic nucleus; mtt = mammillo-thalamic tract; olf-a = olfactory area; olf-n = olfactory nerve; olf =olfactory tubercle; opt = optic tract; pir = piriform cortex; pvs = periventricular system; RF = reticular formation of the brainstem; sa = septal areas; SN = substantia nigra; Sub = subthalamus; zi = zona incerta; III = third ventricle. (From Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.)

Afferents from the brainstem reticular formation include the dorsal longitudinal fasciculus (fasciculus of Schutz), the periventricular fiber system and the medial forebrain bundle.  The dorsolongitudinal fasciculus receives primarily autonomic inputs from centers in the mesencephalic tegmentum (limbic midbrain area), reticular raphe nuclei in the pons and viscero-sensitive nuclei (e.g., the nucleus tractus solitarius) in the medulla oblongata.  The periventricular system carries fibers ascending from both the central grey (including the raphe nuclei) and medial nuclei of the reticular formation in the mesencephalon or dorsal nucleus of the mesencephalic tegmentum (limbic midbrain area). Collectively, these fibers enter the hypothalamus close to the ventricular wall.  The medial forebrain bundle also receives a well-defined fiber tract, the mammillary peduncle, originating from the medial nuclei of the mesencephalic reticular formation (limbic midbrain area).  It courses ventrally in the cerebral peduncle (within the ventral tegmental area of Tsai), and then laterally to the mammillary bodies.

 

Afferents from the thalamus originate in nuclei of the median and medial thalamus, and course in the periventricular system and inferior thalamic peduncle of the ansa pednucularis (an extension of the medial forebrain bundle).

 

Afferents from the subthalamus are believed to originate in the nucleus subthalamicus and zona incerta, and directly enter the hypothalamus along the lateral aspect of the hypothalamic wall (148).

 

Afferents from basal ganglia (corpus striatum) arise from the nucleus accumbens, located in the ventral portion of the caudate nucleus, and via the substantia innominata, directly and indirectly reach the lateral portions of the hypothalamus.

 

Afferents from the retina reach the hypothalamus via the retino-hypothalamic tract, and travel through the optic chiasm to terminate in the suprachiasmatic nucleus.  Finally, direct projections arise from the frontal cortex and course in the medial forebrain bundle to the most lateral part of the ventricular wall.

 

Efferent Connections

 

Outputs from the mammalian hypothalamus include fiber pathways to the anterior and posterior pituitary gland, limbic system, brainstem reticular formation, thalamus, subthalamus, basal ganglia, superior colliculi, substantia nigra, cerebellum, and neocortex (Fig. 23).  With exception of projections to the pituitary gland, discussed in detail below (see Hypothalamic Tuberoinfundibular System and Hypothalamic Neurohypophysial Tract) and those directed to locomotor centers such as the optic tectum, susbstantia nigra, and cerebellum, in general, efferent fibers from the hypothalamus reciprocate its afferent fibers in a sort of feedback loop.

 

Figure 23. Overview of the major efferent pathways from the hypothalamus. (A) Connections with the limbic cortex, brainstem, thalamus and septum. (B) Course of hypothalamic fibers in the dorsal longitudinal fasciculus (fasciculus longitudinalis dorsalis) or fasciculus of Schutz, reaching autonomic and somatic centers in the brainstem and spinal cord. A = anterior hypothalamic nucleus; ac = anterior commissure; cc = corpus callosum; CGRF = central grey - reticular formation; Dm = dorsomedial nucleus; dnmt = dorsal nucleus of the mesencephalic tegmentum; E-Wn = nucleus of Edinger-Wepstal; fb = medial forebrain bundle; ep = epithalamus; fld = fasciculus longitudinalis dorsalis (fasciculus of Schutz); fr = fasciculus retroflexus; ha = habenula; HYP = hypothalamus; htt = habenulo-tectal tract; ilm = intermediate-lateral column of the spinal cord; inf = infundibular or arcuate nucleus; mb = mammillary body; MES = mesencephalon; mfb = medial forebrain bundle; MO = medulla oblongata; mteg = mammillo-tegmental tract; mp = mammillary peduncle; mtt = mammillo-thalamic tract; nts = nucleus tractus solitarius; optec = optic tectum; Po = preoptic area; PP = posterior nuclues; Pv = paraventricular nucleus; pvs = periventricular systems; sm = stria medullaris; so = supraoptic nucleus; RF = reticular formation; rn = red nucleus; tec-spt = tectospinal tract; TS = thoracic spine; Vm = ventromedial nucleus. (From Toni R, Malaguti A, Benfenati F, Martini L: The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.)

Efferents to the limbic system include 2 major fiber groups.  The first course in the medial forebrain bundle (lateral hypothalamus) and carries projections that ascend to the septal areas and descend to the amygdalo-piriform cortex complex.  These latter fibers form the ventral amygdalopetal component of the ansa pedunclaris.  The second, the stria terminalis system with its bed nucleus, is positioned medially, very close to the inner surface of the third ventricle, and projects to the amygdala.

 

Efferents to the brainstem reticular formation include 2 main fiber groups.  The first is a dorsal and medial group formed by the dorsal longitudinal fasciculus and periventricular system.  These axons descend to the brainstem to innervate visceral motor, sensory and somatic nuclei (oculomotor, trigeminal, facial, glossopharyngeal, vagus and accessory spinal nerves), and to autonomic sympathetic and parasympathetic preganglionic neurons in the spinal cord.  The second courses in the medial forebrain bundle and gives off fibers that project to the medial nuclei of the mesencephalic reticular formation (limbic midbrain area) through the mammillary-tegmental tract and the mammillary peduncle.

 

Efferents to the thalamus include 3 main systems.  The first is the mammillo-thalamic tract that links the posterior hypothalamus to the cingulate gyrus of the limbic cortex (a component of the “Papez circuit”).  The second is the periventricular fiber system that projects to the medial (dorsomedian nucleus) and median (midline, intralaminar and reticular nuclei) thalamus and epithalamic habenula through the stria medullaris.  The third is part of the medial forebrain bundle that course in the inferior thalamic peduncle of the ansa peduncularis to reach the medial thalamic nuclei.

 

Efferents to the subthalamic and basal ganglia region course in the lateral aspect of the ventricular wall in the mammillo-thalamic tract (mammillo-subthalamic tract) terminating in the field H1 of Forel, and in the substantia innominata (148,149).  Efferents to superior colliculi, substantia nigra and cerebellum travel in either the stria terminalis and periventricular fiber system, or in the medial forebrain bundle as a part of the mammillary peduncle.  Finally, efferents to the neocortex are carried laterally within the medial forebrain bundle. 

 

Hypothalamic Tuberoinfundibular System

 

The hypothalamic tuberoinfundibular system comprises all neurons in the brain that send axonal projections to the external zone of the median eminence.  Although the arcuate nucleus and inferior portion of the periventricular nucleus were thought primarily responsible for this pathway on the basis of silver stains (150), the relative paucity of myelin in these neurons and high density of perikarya in the medial and periventricular zones of the hypothalamus made it impossible to elucidate the full extent of the tuberoinfundibular system using this technique.  In addition, the inability of pharmacologic ablation of the arcuate nucleus to significantly reduce the concentration of TRH and GnRH in the median eminence (151), made it likely that the origin of neurons contributing to the tuberoinfundibular system is considerably broader than just the medial basal hypothalamus.  Using retrogradely transported marker substances that are taken up in axon terminals in the external zone of the median eminence and transported back to the cell body of origin, a detailed analysis of the tuberoinfundibular system has been possible (152-155).

 

Retrogradely labeled cells of the tuberoinfundibular system concentrate in four major hypothalamic regions: the arcuate nucleus, periventricular nucleus, paraventricular nucleus, and medial preoptic-septal region (Fig. 24).  Within the arcuate nucleus the labeled cells accumulate in two distinct clusters (Fig. 24 G-I), a dorsomedial group of small to medium size neurons in the distribution of the dopamine-containing A12 group of Dahlstrom & Fuxe (156), and a basolateral group of medium sized cells that contain dopamine, GHRH, galanin, galanin-like peptide and neurotensin (156-161).  Occasional enkephalin-producing neurons in the arcuate nucleus are retrogradely labeled from the median eminence (162), but the majority of these cells, as well as ACTH-producing neurons (personal observations), do not contain the retrogradely transported marker substance.  These observations emphasize that only a small subset of chemically coded neurons in the arcuate nucleus project to the median eminence.

 

Figure 24. Coronal sections (rostral to caudal) of rat hypothalamus showing the regional distribution of neurons that accumulate a retrogradely transported marker substance injected into the external zone of the median eminence. Cells of origin of both the tuberoinfundibular pathway and hypothalamic neurohypophysial tract are identified due to diffusion of the tracer into the internal zone of the median eminence. (A-F) Level of the paraventricular nucleus (PVN); (G-I) level of the arcuate nucleus (ARC). III = third ventricle, AH = anterior hypothalamus, M = magnocellular division of the PVN, ME = median eminence, ap = anterior parvocellular subdivision, dp = dorsal parvocellular subdivision, mp = medial parvocellular subdivision, pp = periventricular parvocellular subdivision, vp = ventral parvocellular subdivision of the PVN.

 

In the periventricular nucleus (Fig. 24 A-F), a thin layer of retrogradely marked neurons in the subependymal neuropil contain somatostatin and dopamine (158,163).  These retrogradely labeled cells can be identified even in the most rostral portions of the periventricular nucleus, but the majority concentrate between the middle of the optic chiasm and anterior portion of the median eminence (Fig. 24 B).  Often cells interdigitate between the ependymal wall and even extend into the third ventricular space suggesting possible secretion into the CSF.  Axonal projections from these cells to the median eminence is through a circuitous pathway that extends laterally into the lateral hypothalamus toward the ventral surface of the hypothalamus (retrochiasmatic area) and then medially to enter the median eminence, although some fibers also descend directly in the periventricular neuropil.

 

The most remarkable finding of studies using retrogradely transported marker substances from the median eminence is the massive accumulation of the marker substance in neurons of the paraventricular nucleus (Fig. 24 A-F).  This winged-shaped nucleus at the dorsal margin of the third ventricle can be divided into two major portions based on the size of the neuronal perikarya, including a magnocellular division of large neurons and a parvocellular division of small to medium sized neurons (164).  The parvocellular portion is located in the most medial portion of the nucleus adjacent to the ependymal wall of the third ventricle and can be broken down into several, smaller subdivisions shown in detail in Fig. 25.  Retrogradely labeled cells of the tuberoinfundibular system are located primarily in the anterior, medial and periventricular subdivisions of the paraventricular nucleus with relatively few or no neurons in the dorsal, ventral and lateral parvocellular subdivisions.  Many of these retrogradely labeled cells contain TRH, corticotropin-releasing hormone (CRH), enkephalin, somatostatin, and VIP (162,163,165-169).  Not all neurons in the anterior, medial and periventricular parvocellular subdivisions project to the median eminence, however.  This is particularly apparent for TRH neurons in the anterior parvocellular subdivision that cannot be retrogradely labeled by marker substances introduced into the median eminence (170).  These neurons are also immunocytochemically distinct from hypophysiotropic TRH neurons in the medial and periventricular parvocellular subdivisions in that they do not co-express the peptide, cocaine and amphetamine-regulated transcript (CART) (171).  The true, physiologic function of TRH neurons in the anterior parvocellular subdivision is not known.

 

Figure 25. Schematic of the hypothalamic PVN showing major subdivisions. (A) Anterior, (B) Mid, and (C) Caudal levels. AP = anterior parvocellular subdivision, DP = dorsal parvocellular subdivision, LT = lateral parvocellular subdivision, MN = magnocellular division, MP = medial parvocellular subdivision, P = periventricular parvocellular subdivision, VP = ventral parvocellular subdivision.

Tuberoinfundibular neurons in the paraventricular nucleus project to the median eminence either by arching laterally and inferiorly through the lateral hypothalamus through the retrochiasmatic area before turning medially to terminate or by descending along the wall of the third ventricle to directly enter the median eminence.  As the ependymal wall underlies both the paraventricular and periventricular nuclei and is likely permeable to the diffusion of CSF (68), these cells of the hypothalamic tuberoinfundibular system could be influenced by substances carried in the CSF or secrete directly into the CSF as an alternative way to reach the median eminence.

 

Finally, small, bipolar and multipolar, retrogradely labeled cells that can be immunostained with GnRH (169,172), are found in the rostral hypothalamus in the ventral wings of the diagonal band of Broca, lamina terminalis, medial septum, and medial preoptic nucleus, while few cells extend more caudally in the basolateral hypothalamus.  In primates, however, retrogradely labeled GnRH cells are located more caudally in the basal hypothalamus (173).  Axonal projections to the external zone of the median eminence occur either by joining the medial forebrain bundle in the lateral hypothalamus or along the wall of the third ventricle.  The tendency for GnRH neurons of the tuberoinfundibular pathway to be more deeply embedded into the substance of the hypothalamus than is typical for the periventricular distribution of the majority of the hypothalamic tuberoinfundibular system relates to the embryologic origin of GnRH neurons from the nasal epithelium (174), as opposed to primordial cells in the walls of the third ventricle.

 

Although the bulk of tuberoinfundibular neurons arise from periventricular and medial portions of the hypothalamus, some brain stem neurons also have direct projections to the median eminence, explaining the presence of catecholamines in addition to dopamine in this structure.  Retrogradely neurons can be identified in C1-C2 adrenergic neurons and A2 noradrenergic neurons (175), but since the tracer was injected into the bloodstream in this study, uptake could have occurred from other circumventricular organs in addition to the median eminence.  Lesions of the brainstem, however, do result in degeneration of axon terminals in the median eminence (176).

 

Hypothalamic Neurohypophysial Tract

 

The hypothalamic neurohypophysial tract defines the neuronal system terminating in the posterior pituitary and is best known for its secretion of vasopressin and oxytocin into the peripheral circulation to regulate water balance (antidiuresis), milk ejection and uterine contraction (177).  Neurons of this tract arise primarily from the magnocellular division of the paraventricular nucleus and the supraoptic nucleus (178), the latter situated as a cluster of cells dorsal and lateral to the optic chiasm (Fig. 26).  The axon trajectory from magnocellular neurons to the posterior pituitary is by way of arching fibers extending laterally and inferiorly from the paraventricular nucleus above and below the fornix toward the supraoptic nucleus, where it gathers fibers from the supraoptic nucleus and continues medially along the base of the hypothalamus into the internal zone of the median eminence.  Vasopressin-containing axon terminals have also been demonstrated in the external zone of the median eminence, particularly following adrenalectomy (179), but largely arise from a separate population of parvocellular neurons in the paraventricular nucleus that contain CRH (180).  Vasopressin is a weak corticotropic factor but potentiates the secretion of ACTH in the presence of CRH (181) and is responsible for the ACTH rise following hypoglycemia (182).

Figure 26. Organization of the hypothalamic neurohypophysial tract (arrows). Note arching fibers emanating from magnocellular neurons in the paraventricular nucleus (PVN) as they descend toward and join fibers emanating from the supraoptic nucleus (SON). The fiber tract converges in the midline at the base of the hypothalamus in the retrochiasmatic area (arrowheads) before entering the internal zone of the median eminence. III = third ventricle, F = fornix, OC = optic chiasm.

Magnocellular neurons of the paraventricular and supraoptic nucleus possess large perikarya and prominent dendrites that interdigitate with adjacent perikarya and dendrites, respectively, of other magnocellular neurons.  These dendrites contain numerous hormone-laden neurosecretory granules that can be released by exocytosis (183) and may be important to coordinate the secretion of vasopressin or oxytocin from individual neurons in unison through somato-somatic and/or dendro-dendritic interactions, or alter the sensitivity of these neurons in response to a biologic stimulus such as suckling.  Regulation of magnocellular neurons may also depend upon dynamic glial-neuronal interactions in response to specific stimuli, reducing or enlarging the cell to cell contact area between magnocellular neurons by retraction or extension of astrocytic processes that separate perikarya and dendrites (184), or to permit the formation of new synaptic contacts on magnocellular neurons (synaptic plasticity) (185).

 

In addition to vasopressin and oxytocin, magnocellular neurons of the hypothalamic neurohypophysial tract also produce and transport numerous other peptides to the posterior pituitary.  Coexisting in vasopressin axon terminals are dynorphin, enkephalin, galanin, cholecystokinin, dopamine, TRH, VIP, neuropeptide Y, substance P, CRH, endothelin, pituitary adenylate pituitary cyclase-activating polypeptide (PACAP), secretin and glutamate, and in oxytocin terminals, dynorphin and proenkephalin A-derived µ-opioid peptides (186-190).  A number of different neuropeptides are also carried into the posterior pituitary by axons from parvocellular neurons including GnRH, TRH, somatostatin, enkephalin, neurotensin CRH and dopamine (191).  Furthermore, there is evidence that messenger RNA for vasopressin, oxytocin (192) and tyrosine hydroxylase (193) can be transported in axons of the hypothalamic neurohypophysial tract, particularly during osmotic stress.

 

The functional significance of numerous biologically active substances in axons from both magnocellular and parvocellular neurons, other than vasopressin and oxytocin, is of great interest.  Since their concentration in the posterior pituitary is low, release into the peripheral circulation for action at a distant locus seems remote.  Endothelin-1 may be an exception, however, where co-release with vasopressin into the periphery may assist the effect of vasopressin on water conservation by decreasing glomerular filtration rate (157).  Other substances are likely involved in the regulation of vasopressin and oxytocin secretion by a paracrine or autocrine mechanism or by acting presynaptically on nerve endings in the posterior pituitary.  Dopamine, for example, may be important in stimulating vasopressin release during an osmotic challenge (194).  Neuropeptide Y has also been shown to enhance the secretion of vasopressin (195) and NPY Y2 receptors are present on nerve endings in the posterior pituitary in high density (196).  Some neuropeptides in the posterior pituitary may act as trophic hormones (98), important to promote regeneration of its axon terminals following injury or to increase the proliferation of endothelial cells to promote changes in its vascularization (197).

 

There is considerable evidence, however, that some of the peptides in the posterior pituitary are destined for transport to the anterior pituitary via the short portal vessels, thereby utilizing the posterior pituitary as an accessory median eminence.  The most convincing data that the posterior pituitary can influence anterior pituitary function comes from animal studies in which the posterior pituitary is surgically removed.  Consequently, the blood flow to the anterior pituitary from the median eminence through the long portal veins is preserved, but the blood flow from the posterior pituitary through the short portal veins is disrupted.  These animals have a number of abnormal neuroendocrine responses including a diminished ACTH response to stress (198), elevation in basal prolactin levels (193), and the loss of either suckling or estrogen induced prolactin release (199,200).  These responses indicate the requirement of magnocellular derived-vasopressin or some other posterior pituitary secretagogue (CRH, oxytocin, dopamine, TRH, other prolactin releasing factor) to achieve normal physiologic responses.

 

Radiologic Anatomy

 

Magnetic resonance imaging (MRI) gives remarkable detail of the hypothalamus (Fig. 27) and thereby, has become the major radiologic tool to assess pathology in this region of the brain (201).  While individual hypothalamic nuclear groups cannot be identified with this technique, some of the major fiber tracts that traverse the hypothalamus can be seen as high intensity signals, particularly in T-2 weighted images (202).  These tracts include the fornix and the mammillothalamic tract, shown in Fig. 27B.  Thus, using these fiber pathways as anatomical landmarks, it is possible to radiologically divide the hypothalamus into the two major subdivisions, the medial and lateral hypothalamic areas. In addition, in the most rostral portions of the hypothalamus, the anterior commissure is readily seen by MRI (Fig. 27A), and increased signal in the lateral hypothalamus is most likely due to the presence of the medial forebrain bundle (Fig. 27B-D).

 

Figure 27. MRI of coronal sections through the hypothalamus. (A) Anterior hypothalamus corresponding to Fig. 16A showing location of the anterior commissure (arrows). (B) Mid hypothalamus corresponding to Fig. 16B showing location of the fornix (arrow). (C) Mid hypothalamus corresponding to Fig. 16C showing the optic tract. The fornix can sometimes also be visualized at this level. (D) Caudal hypothalamus corresponding to Fig. 16D at the level of the medial mammillary bodies (arrow). Sometimes the mammillothalamic tract can be visualized at this level.

 

Blood Supply

 

All arteries carrying blood to the hypothalamus are terminal branches of the circle of Willis, including the internal carotid, anterior cerebral, anterior communicating, posterior communicating, posterior cerebral and basilar arteries (Fig. 13).  Curiously this arterial circle, named for Thomas Willis’s work published in Cerebri Anatome in 1664 (Fig. 28), had already been described between the end of the 16th century and beginning of the 17th century by the Italian anatomists, Fallopius and Casserio (203).  These anatomists noted the existence of small arterial branches entering the floor of the third ventricle and surrounding the tuber cinereum in a position that is now well described as the anastomotic circuminfundibular plexus and prechiasmal anastomotic arteriolar-capillary plexus.  These two anastomotic plexuses are highly developed in species such as Carnivora, Cetacea, Edentata and Ungulata (204) and likely represent the capillary system that Galen described as the rete mirabilis, given that he worked on animal species and not human brains and the anatomical differences between what he described and what is now recognized as the primary portal plexus (see below).

 

Figure 28. Original depiction of the arterial circle (or “polygon”) surrounding the hypothalamic infundibulum at the base of the brain, as shown in the Cerebri Anatome (1664) by Thomas Willis and drawn by the British architect, Cristopher Wren. Note the presence of branches to the mammillary bodies but absence of vessels to the infundibulum. (Courtesy of the Library of the Department of Human Anatomy of the University of Parma, Italy.)

The vascular supply to the hypothalamus has been extensively studied in man using three-dimensional casts of the hypothalamic vessels (205).  These studies demonstrate that the arterial supply is compartmentalized with respect to three rostro-caudal regions of the hypothalamus, and thus, can be separated into anterior, intermediate and posterior arterial groups (Table 8).  However, only the anterior hypothalamic region is vascularized by a single arterial group (the anterior arterial group), whereas the remainder of the hypothalamus receives blood from both the intermediate and posterior arterial groups.  The preoptic and anterior hypothalamus are primarily supplied by the anterior cerebral and anterior communicating arteries, the tuberal region by the posterior communicating artery, and the mammillary region by the posterior communicating, posterior cerebral, and basilar arteries.  This organization is consistent with the clinical observations that occlusion of the anterior choroidal artery, which has anastomotic branches with the posterior communicating artery, results in damage to the tuberal and mammillary regions of the hypothalamus, whereas occlusion of the thalamoperforate artery, a branch of the posterior cerebral artery, results in damage to the mammillothalamic tract and related thalamic nuclei (205).

 

Table 8.  Arterial Groups that Supply Hypothalamic Nuclei

ANTERIOR GROUP

internal carotid, anterior cerebral and posterior communicating arteries

INTERMEDIATE GROUP

posterior communicating artery

POSTERIOR GROUP

posterior communicating, posterior cerebral and basilar arteries

AHA

ARC

LHA

MPA

DMN

LMN

PV

LHA

MMN

PVN

LMN

PN

SCN

LTN

PV

SON

MMN

SMN

 

PN

 

 

PV

 

 

SMN

 

 

VMN

 

AHA = anterior hypothalamic area, ARC = arcuate nucleus, DMN = dorsomedial nucleus, LHA = lateral hypothalamic area, LMN = lateral mammillary nucleus, LTN = lateral tuberal nucleus, MMN = medial mammillary nucleus, MPA = medial preoptic area, PN = posterior hypothalamic nucleus, PV = periventricular nucleus, PVN = paraventricular nucleus, SCN = suprachiasmatic nucleus, SMN = superamammillary nucleus, SON = supraoptic nucleus, VMN = ventromedial nucleus

 

As previously described, the blood supply to the median eminence is complex.  In all mammals, including humans, fine branches of the superior hypophysial artery give rise to a capillary plexus, the primary portal plexus of the infundibulum, which is composed of capillary loops in the external zone of the median eminence (external plexus) and that penetrate as pillars vertically toward the ventricular floor to establish the internal plexus.  At this level, a subependymal capillary network can also be recognized in association with the basal membrane of the ependymal cells.  Blood flows from the external to the internal and then back to the external plexus to end in the long portal vessels that reach the anterior pituitary, or from the subependymal network into hypothalamic capillaries of the anterior or intermediate arterial groups (206).  In some species, bidirectional transport of substances has been described in the portal capillary system, allowing the transport of anterior pituitary substances to the external plexus (207), thus supporting the original hypothesis of Popa and Fielding that in the human brain, blood flow in the portal capillary system can be from the pituitary to the hypothalamus (208).  The external plexus is tangential to the ventral surface of the infundibulum and is composed of vessels organized in geometrical arrays (hexagonal in rodents, much more complex in humans), whose central spaces are filled with neuroendocrine axons constituting functional units, called microvascular domains (206) or medianosomes (209).  Neurohemal contacts are established by these axons and by tanycyte processes at the level of both plexuses, whereas neurohypophysial fibers course between the two vascular plexuses without contacting them, en route to the posterior lobe of the pituitary.  This “double-plexus” system provides amplification of the surface area in contact with tuberoinfundibular axons in a given microvascular domain.

 

Venous drainage from the rest of the hypothalamus is collected into the anterior cerebral, basal, and the internal cerebral veins, ultimately reaching the great vein of Galen.  In general, the anterior cerebral and basal veins drain the majority of the hypothalamus, whereas the internal cerebral vein collects blood from the dorsal portions of the hypothalamus (Table 9).

 

Table 9.  Venous Drainage from Hypothalamic Nuclei

Anterior Cerebral and Basal Veins

Internal Cerebral Vein

AHA

DMN

ARC

LHA

LHA

LMN

LMN

MMN

LTN

PV

MMN

PVN

MPA

SMN

PN

 

PVN

 

SCN

 

SMN

 

SON

 

VMN

 

AHA = anterior hypothalamic area, ARC = arcuate nucleus, DMN = dorsomedial nucleus, LHA = lateral hypothalamic area, LMN = lateral mammillary nucleus, LTN = lateral tuberal nucleus, MMN = medial mammillary nucleus, MPA = medial preoptic area, PN = posterior hypothalamic nucleus, PV = periventricular nucleus, PVN = paraventricular nucleus, SCN = suprachiasmatic nucleus, SMN = superamammillary nucleus, SON = supraoptic nucleus, VMN = ventromedial nucleus

 

Glymphatic System and Dural Sinus-associated Lymphatics

 

Recent studies in rodents have revealed that fluorescent tracers injected into the arachnoid cisternae can travel along a perivascular network of channels coursing throughout the brain, the hypothalamus included, and contain cerebrospinal fluid (CFS), water, food metabolites (primarily lipids) and proteins arising from tissue degradation/inflammation (e.g., β-amyloid), coined the glymphatic system (210).  Although these findings replicate similar observations made more than 30 years ago using, horseradish peroxidase in dogs and cats (211), it is only recently that it became clear that these channels do originate as a continuation of the subarachnoid cisternae around the arteries dividing inside the brain parenchyma otherwise known as Virchow-Robin spaces.  Virchow-Robin spaces are externally lined by the glia limitans that gives rise to a pavement underneath the basal lamina of the perivascular leptomeningeal cells facing the brain parenchyma, whereas a symmetrical leptomeningeal lining embraces the basal lamina of the smooth muscle, arterial cells.  Once the arteries enter the depth of the brain matter to become penetrating arterioles and capillaries, they lose both their muscular sheath and perivascular leptomeninges, but a narrow interstitum persists between the basal lamina of the vascular endothelial cells and that of the original subarachnoid cells.  The latter basal lamina is ensheated by the endfeet of aquaporin4 (AQP4)-expressing astrocytes that form the external boundaries of the glymphatic interstitium, where they regulate the passage of water and intercellular solutes from the interneuronal spaces into the perivascular CSF, and vice versa.  The absence of endothelial tight junctions in circumventricular organs (63) such as the median emince, amplifies the opportunity for reciprocal exchange with blood-born substances.  The liquid bulk is passed into a connected perivenular and perivenous space (212) that then drains into the extracranial perivenous lymphatics (213) (Fig. 29).

Figure 29. Composite of different drawings schematizing the 3D organization of the brain glymphatic system with particular reference to the hypothalamic-pituitary unit. A) Distribution by magnetic resonance imaging of paramagnetic contrast (yellow arrows) in paravascular channels of the rat hypothalamus (HYP), pituitary (Pit), and pineal (Pin) recesses following injection into the cisterna magna (adapted from ref. 30). A contingent of glymphatic flow connects the medial-basal hypothalamus to the limbic structures and olfactory area (OA) and lobe (OL), ensuring a wide interplay by local soluble materials (volume transmission); B) Structure of the glymphatic channels (adapted from ref. 212): pial arteries (Ar) coursing in the subarachnoid space are surrounded by CSF that flows in the perivascular space, termed the Virchow–Robin space. The arteriolar tunica media is isolated from the CSF by a lining of leptomeningeal cells; however, the CSF-containing Virchow–Robin spaces progressively narrow and finally disappear at the level of capillaries (red rectangle). The CSF continues its flow into perivascular spaces provided only by the extracellular matrix of the basal lamina. Astrocytic vascular endfeet expressing aquaporin-4 (AQP4) surround the entire vasculature and form the boundary of the perivascular spaces. These astrocytes may transfer information carried by solutes in the glymphatic system to surrounding glial cells and neurons, and vice versa; C) the glymphatic spaces are in continuity with venules and veins (adapted from ref. 210), allowing for glymphatic drainage either into sinus-associated dural lymphatics and venous brain sinuses or exiting the brain into perivenular lymphatics and lymph nodes.

Remarkably, the fluid dynamics in the hypothalamic glymphatic channels is affected by alterations in carbohydrate metabolism, such as diabetes mellitus, as a result of enlargement of the paravascular spaces by structural arteriolar and/or venular damage (214).  This suggests a potential role for the hypothalamic glymphatic network in regulating the intrahypothalamic exposure of glucose, free fatty acids, and proteins to relevant hypothalamic nuclei involved in the regulation of appetite and satiety (See section on Appetite and Satiety). Similar, transthyretin-coupled thyroid hormones reaching the CSF from the choroid plexus (215) might enter the hypothalamic glymphatic system and act as an amplifier of a thyroid-dependent, volume transmission in the hypothalamus (216), influencing a large array of hypothalamic homeostatic neurons (see section on Functional Anatomy of Hypothalamic Homeostatic Systems).

 

The glymphatic channels are in position to merge with dural lymphatics.  The latter were originally described in the dura mater and pia mater in human cadavers in 1787 by the Italian anatomist, Paolo Mascagni (217), and recently rediscovered in rodent models (218,219) as an unexpected anatomical novelty (220), challenging the concept of brain parenchyma as a site of “immune privilege”.  Endothelial cells of these conduits express markers of the lymphatic vessels including LYVE1, PROX1 and VEGFR3, and course parallel to both the main dural sinuses (sagittal and transverse) and dural middle meningeal arteries, the latter laying adjacent to the dural boundaries of the cavernous sinus where dural lymphatics might establish a direct cavernous connection (30). Therefore, it has been suggested that the perivascular pathways of the glymphatic system and meningeal lymphatic vessels should be considered as serial elements of an interconnected circuitry for immune surveillance of the brain by the immune system, presumably allowing brain antigens access to lymph nodes outside the brain parenchyma (221).  Such a circulatory pathway may contribute to the pathogenesis of inflammatory and autoimmune conditions of the hypothalamic-pituitary unit (222,223) including lymphocytic hypophysitis and diabetes insipidus (Fig. 29).

 

FUNCTIONAL ANATOMY OF HYPOTHALAMIC HOMEOSTATIS SYSTEMS

 

Regulation of Hypophysiotropic Neurons

 

The secretion of hypothalamic releasing and inhibitory hormones from axon terminals of tuberoinfundibular neurons into the portal capillary system is dependent upon several layers of control that can be exerted directly on the perikarya and/or processes of these neurons.  For one, neurons of the tuberoinfundibular system can be modulated by substances circulating in the bloodstream that either pass the blood-brain barrier because they are fat soluble steroids or small molecules, or access tuberoinfundibular neurons via the cerebrospinal fluid due to the periventricular location of many tuberoinfundibular neurons and poor development of tight junctions between ependymal cells in these regions (63).  Feedback effects of thyroid hormone, for example, occur directly on TRH-producing neurons within the paraventricular nucleus as demonstrated by the ability of a microcrystalline implant of T3 adjacent to the paraventricular nucleus to prevent the hypothyroid-induced increase in TRH biosynthesis on that side but not the opposite side (224,225).  In addition, tuberoinfundibular neurons receive numerous axosomatic and/or axodendritic contacts from local interneurons and/or other regions in the brain that contain a variety of chemical messengers that contribute to intercommunication between specific neuronal groups or are important in establishing the set point at which the hypophysiotropic substances are secreted in response to hormonal feedback signals.  To demonstrate how the CNS can exert regulatory control over hypophysiotropic neurons, examples of modulation of GH secretion and regulation of the hypothalamic-pituitary-adrenal (HPA), hypothalamic-pituitary-thyroid (HPT), and reproductive axes are given below.

 

MODULATION OF GHRH/SRIF TUBEROINFUNDIBULAR NEURONS

 

A well-studied example of local afferent influences on the activity of tuberoinfundibular neurons is demonstrated by the hypothalamic regulatory system involved in the control of GH secretion.  The pattern of GH secretion is episodic, showing a regular periodicity of one pulse every 2 to 4 hours and low or undetectable trough values (226).  This rhythm is the result of the control by two separate components of the tuberoinfundibular system, including GHRH-producing neurons (stimulatory) in the basolateral portion of the arcuate nucleus and somatostatin-producing neurons (inhibitory) in the periventricular nucleus, each secreting into the portal capillary plexus.  To coordinate this rhythmic secretion, reciprocal axonal connections between these two populations of neurons may be necessary (Fig. 30).  In this manner, somatostatin neurons receive direct, stimulatory inputs from GHRH neurons while GHRH neurons receive direct, inhibitory inputs from somatostatin-containing neurons, Treatment of hypothalamic cultures with somatostatin inhibits GHRH, whereas treatment with GHRH induces somatostatin release (227).  In addition, both GH and IGF-1 cross the blood-brain barrier with GH increasing somatostatin secretion into the portal capillary system, whereas IGF-1 both increases somatostatin and inhibits GHRH, contributing to a finely tuned regulatory system (228,229).  GH secretion can also be modulated by a number of other factors including glucose and free fatty acids circulating in the bloodstream, but also neurotransmitters and peptides intrinsic to the central nervous system, the latter acting primarily on somatostatin and/or GHRH producing tuberoinfundibular neurons.  For example, GH receptors are expressed by the majority of NPY neurons in the hypothalamic arcuate nucleus that show c-Fos expression in response to GH administration and are known to innervate periventricular somatostatin neurons and increase somatostatin secretion (230).  The rise of GH during sleep is probably mediated by cholinergic pathways suppressing somatostatin secretion (231).  Stress and sepsis can also be associated with a rise in GH levels mediated by catecholamines by increasing GHRH (232).  The precise origin of the neurons giving rise to these neuromodulators, however, is not known.  Glutamate may also be of importance by increasing the secretion of GHRH as N-methyl-D-aspartate increases GH secretion (233), and this can be attenuated by GHRH antibodies (234).  In addition, the majority of GHRH neurons in the arcuate nucleus are contacted by axons expressing the glutamate transporter 2 (VGLUT2) (235), a selective marker for glutamatergic elements.  PACAP also increases GH secretion, but PACAP knock-outs do not have disturbance in GH release (236,237).  Multiple other peptides have also been shown to have either a stimulatory or inhibitory effect on GH secretion but their physiological significance is unknown (238).

Figure 30. Schematic representation of the interactions between somatostatin (SRIH)-producing neurons in the hypothalamic periventricular nucleus (Pev) and growth hormone releasing hormone (GHRH)-producing neurons in the arcuate nucleus. Note that in addition to projections to the external zone of the median eminence, these neurons may also have reciprocal connections. Ghrelin (GHR) may also influence GH secretion by acting directly on somatotrophs or through stimulatory effects on GHRH neurons. AP = anterior pituitary, GH = growth hormone, IGF = insulin-like growth factor, ME = median eminence, PP = posterior pituitary. R-SRIH and R-GHRH correspond to receptors for the respective peptides. (Adapted from Epelbaum J: Intrahypothalamic neurohormonal interactions in the control of growth hormone secretion. Functional Anatomy of the Neuroendocrine Hypothalamus, Wiley, Chichester, 1992; pp 54-68.)

A relatively new and potentially exciting chapter in the understanding of the physiology of GH secretion has been the discovery of ghrelin, the most potent (on a molar basis) GH secretagogue known to man (239).  Ghrelin circulates in the bloodstream, secreted primarily from the stomach, but is also produced by neurons in the hypothalamic arcuate nucleus (240).  When administered as a single intravenous dose, it induces an acute release of GH, and when administered continuously, it increases 24h pulsatile secretion of GH (241).  Although the anterior pituitary somatotrophs contain receptors for ghrelin, and ghrelin can directly induce GH secretion, it is more likely that ghrelin exerts its main effects in the hypothalamus by triggering GHRH secretion (242), as pulsatile patterns of plasma or CSF ghrelin do not correlate well to circulating GH concentrations (243).   Ghrelin levels fall in response to rising levels of GH, providing evidence for a gastro-hypophysial feedback loop (244).  Other external and metabolic signals also can modulate GH secretion such as the decrease in blood glucose following a carbohydrate-rich meal, hypoglycemia, and a high intake of protein that are believed to function through a common mechanism of somatostatin withdrawal and disinhibition of GH secretion (245).

 

MODULATION OF CRH TUBEROINFUNDIBULAR NEURONS

 

Analogous to the hypothalamic-pituitary-thyroid axis alluded to above and discussed in greater detail in the following section, the hypothalamic-pituitary-adrenal axis is similarly modulated by direct feedback effects on hypophysiotropic CRH neurons by a circulating hormone, in this case glucocorticoids. Removal of the adrenals results in marked upregulation of CRH mRNA selectively in the PVN.  Glucocorticoids can also exert additional, rapid signaling on hypophysiotropic CRH neurons via endocannabinoid-mediated suppression of synaptic excitation, in which 2-arachidonooyl-glycerol [2-AG] and N-arachidonoylethanolamine [AEA] from CRH neurons inhibit the release of glutamate from neurons synapsing on hypophysiotropic CRH neurons (246).

 

Afferent input to tuberoinfundibular neurons from distant loci in the brain, howere, is another important regulatory mechanism over hypophysiotropic function and is one way that tuberoinfundibular neurons are integrated with other functions of the brain.  The parvocellular subdivision of the paraventricular nucleus receives direct, dense, afferent input from autonomic centers in the lower brain stem including the nucleus of the tractus solitarius (NTS), dorsal motor nucleus of the vagus (DMNv) and several catecholamine groups in the dorsal and ventral lateral medulla, carrying visceral sensory information primarily from the abdomen and thorax through the vagus and glossopharyngeal nerves (247).  At least part of this projection is noradrenergic but other substances such as neuropeptide Y (248), activin (249), and GLP-1 (250) are also carried in these fibers, some coexisting in catecholaminergic neurons.  After traversing through the medial forebrain bundle in the lateral hypothalamus, axons containing catecholamines have been observed to make numerous synaptic contacts with CRH-producing neurons in the paraventricular nucleus (251,252) and to induce the secretion of CRH primarily via α1 adrenergic receptors (253).  In this manner, sensory information from the periphery (e.g., heart rate, blood pressure as during hemorrhagic shock) has the potential to alter the set point for the secretion of hypophysiotropic CRH using norepinephrine as the central mediator, and thereby increase circulating levels of glucocorticoids.

 

In a similar fashion, increased secretion of glucocorticoids in response to infection or inflammation is due to the activation of catecholaminergic neurons of the NTS (A2 noradrenergic, C2 adrenergic) and rostral ventrolateral medulla but initiated by endotoxin and proinflammatory cytokines such as interleukin-1 (IL-1) (254).  Under these circumstances, the set point for feedback inhibition of hypophysiotropic CRH secretion is altered to allow the powerful immunosuppressant action of glucocorticoids to limit the severity of the inflammatory response (255).  If the ascending catecholamine pathway to the PVN is transected, the ability of IL-1 to increase CRH mRNA in PVN neurons is reduced (256).  It is proposed that IL-1 exerts its effect on endothelial cells and/or perivascular microglia at the blood-brain interface, resulting in activation of cyclooxygenase-2 (the rate limiting enzyme for the formation of prostaglandins), the release of prostaglandin E2 (PGE2) into the surrounding tissue, and ultimately activation of catecholaminergic neurons through prostaglandin receptors (257).   This hypothesis is supported by the demonstration that focal injection of PGE2 into the medulla reproduces the activating effects of IL-1 on CRH neurons in the PVN (258).  Alternatively, cytokines may exert their effects on vascular cells at the blood-brain-barrier directly within the PVN, itself, or after penetrating the blood-brain barrier at circumventricular organs such as the OVLT, and then transmitting the information through neural pathways that interconnect these structures with the PVN (257, see Thermoregulation below).

 

Neurogenic stress also leads to resetting of the HPA axis and similarly characterized by elevated circulating glucocorticoid levels and increased CRH gene expression in hypophysiotropic neurons (254).  However, the mechanism would appear to be vastly different than that described above as indicated by persistent HPA activation under these conditions despite disruption of the ascending catecholaminergic pathways to the PVN (259).  CRH neurons receive inputs from other portions of the brain such as the forebrain limbic system, and surgical ablation of hippocampal efferents to the hypothalamus (260) or lesions in the bed nucleus of the stria terminalis (261) increase the concentration of CRH mRNA in the paraventricular nucleus.  In addition, activation of NPY neurons originating in the hypothalamic arcuate nucleus may contribute to increased CRH cellular activity induced by insulin hypoglycemia (262,263).  Interactions among axon terminals containing GABA, glutamate and endocannabinoids may also contribute to modulation of stress-induced activation of CRH neurons (264).  Thus, while the end result to increase circulating levels of glucocorticoids is similar in all stress paradigms, depending upon the type of stress, different regions of the brain are recruited to allow resetting of the HPA axis.  The mitogen activated protein (MAP) kinase signaling pathway may be a common final mechanism in hypophysiotropic CRH neurons that links a variety of different stimuli that activate the HPA axis to an increase in CRH gene expression (265-267).  Stress and glucocorticoid-induced synaptic plasticity of the neural circuitry regulating hypophysiotropic CRH neurons may also contribute to regulation of the HPA axis, particularly in the face of chronic stress (264).

 

The circuitry described above that allows resetting of the HPA axis is illustrated in Fig. 31.

 

Figure 31. Schematic diagram showing autonomic regulation of corticotropin-releasing hormone (CRH) neurons of the hypothalamic tuberoinfundibular tract. (1) Visceral sensory information carried by the glossopharyngeal (IX) and vagus (X) nerves terminates in lower brain stem autonomic centers which (2) project to CRH neurons in the paraventricular nucleus (PVN), (3) modulating the secretion of CRH into the portal capillary system in the median eminence (ME). Descending pathways from the PVN reach autonomic centers in the (4) lower brainstem and (5) spinal cord and can influence the autonomic nervous system. (6) CRH neurons also receive direct input from the limbic system via bed nucleus of the stria terminalis. IML = intermediolateral cell column of the spinal cord. (Schematic diagram based on rat data and modified from Sawchenko PE, Swanson LW, Science 1981; 214:685-687.)

MODULATION OF TRH TUBEROINFUNDIBULAR NEURONS

 

The secretion of hypophysiotropic TRH is primarily regulated by negative feedback regulation by thyroid hormone mediated by the beta 2 isoform of the thyroid hormone receptor (TRβ2) expressed on these neurons (268).  As T3, alone, is not sufficient to normalize TSH expression in the PVN except when administered in thyrotoxic doses (269), under normal conditions, feedback inhibition is dependent upon the conversion of T4 to T3.  Hypophysiotropic TRH neurons, however, do not contain the enzyme necessary for T4 to T3 conversion, type 2 iodothyronine deiodinase (D2).  Rather, it is believed that T3 production is mediated by tanycytes, specialized ependymal cells lining the third ventricle.  These cells not only contain D2, but also express the thyroid hormone transporters, OATP1 and MCT8, on their surface.  It is proposed that T3 released from tanycytes contribute to feedback regulation by thyroid hormone viamechanisms further elaborated below (270).  As noted previously, tanycytes also express the TRH degrading ecto-enzyme, pyroglutamyl peptidase II (PPII), in keeping with the important role of tanycytes in control of the hypothalamic-pituitary-thyroid axis.  Hypophysiotropic TRH neurons also express the vesicular glutamate transporter 2 (VGLUT2), whereas tanycytes the line the median eminence express AMPA and kainite receptors and glutamate transporters and are depolarized by glutamate and by optogenetic activation of TRH axons in the median eminence (271).  As the majority of hypophysiotropic TRH neurons express the type 1 cannabinoid receptor (CB1) mRNA and contain punctate CB1-immunoreactive signal in their axon varicosities, tanycytes in close association with TRH-containing axon terminals in the median eminence contain the endocannabinoid synthesizing enzyme, diacylglycerol lipase α (DAGLα), antagonizing CB1 or inhibiting DAGLα stimulates TRH release in median eminence explants and inhibition of glutamate signaling markedly decreases the 2-AG content of the median eminence (271), the existence of a microcircuit in the median eminence is suggested in which the activity of TRH axons stimulate endocannabinoid synthesis in tanycytes, whereas the tanycytes restrain the delivery of TRH to the anterior pituitary via the endocannabinoid system.  This interaction may have importance in the regulation of pulsatile secretion of TRH and provide and extra flexibility to the control of TRH release.

 

Elucidation of the mechanisms by which the hypothalamic-pituitary-thyroid (HPT) axis responds to fasting provides another excellent example of how afferent input from neurons arising outside of the PVN can influence the secretion of hypophysiotropic neurons.  Similar to the feedback mechanisms controlling the adrenal axis, maintenance of normal thyroid function is dependent upon a negative feedback control system.  Circulating levels of thyroid hormone (T4 and T3) influence the biosynthesis and secretion of TRH in hypophysiotropic neurons in the PVN (Fig. 32) and TSH in the anterior pituitary (225), although a role for thyroid hormone-induced upregulation of the TRH degrading enzyme, pyroglutamyl peptidase II, expressed in tanycytes has also been proposed by regulating the amount of TRH released into the portal system (272).  In response to fasting or infection, however, this normal homeostatic system is altered in a way that is presumably beneficial for survival.  Under these circumstances, there is a fall in circulating thyroid hormone levels but a seemingly paradoxical reduction in TRH gene expression in the PVN (Fig. 33), reduced secretion of TRH into the portal blood and low or inappropriately normal plasma TSH (273-276), rather than the anticipated increase in all of these parameters as seen in primary hypothyroidism mediated by a decline in circulating levels of leptin (see below for mechanism).  Thus, during fasting, the normal feedback mechanism described above is overridden, and a state of central hypothyroidism is transiently induced.  Presumably, by reducing thyroid thermogenesis and preserving nitrogen stores, this mechanism is an important adaptive response to reduce energy expenditure until the adverse stimulus is removed (277).

Figure 32. High magnification in situ hybridization autoradiographs of proTRH mRNA in the paraventricular nucleus (PVN) of a (A) euthyroid and (B) hypothyroid animal. Note marked increase in TRH mRNA when circulating levels of thyroid hormone fall.

The HPT axis is primarily modulated by afferent input derived from the hypothalamus, itself.  At least two anatomically distinct populations of neurons in the arcuate nucleus with opposing functions, proopiomelanocortin (POMC)-producing neurons that also co-express cocaine and amphetamine-regulated transcript (CART), and NPY-producing neurons that co-express agouti related peptide (AGRP), appear to be responsible (174,278-280).  Both neuronal populations express receptors for the white adipose tissue-derived circulating hormone, leptin, and project to hypophysiotropic TRH neurons in the PVN through a monosynaptic, arcuate-PVN pathway (281-283).  Alpha-MSH, a translation product of POMC, and CART (originally described as a mRNA induced in the striatum following psychostimulant drug administration) both induce transcription of the TRH gene in hypophysiotropic neurons (163,278), whereas NPY and AGRP are inhibitory (279,280), NPY via direct effects on Y1 and Y5 receptors on TRH neurons (269), and AGRP by antagonizing α-MSH at melanocortin receptors (285).  Thus, during fasting when circulating levels of leptin decline, expression of the genes encoding POMC and CART are reduced simultaneously with a marked increase in the genes encoding NPY and AGRP (286,287), effectively lowering the threshold of feedback inhibition of hypophysiotropic TRH by circulating levels of thyroid hormone.

Figure 33. In situ hybridization autoradiographs of proTRH mRNA in the PVN (arrow) of (A) normal fed and (B) fasting animals. Note the marked reduction in hybridization signal by fasting. (C) ProTRH mRNA levels are restored to normal in fasting animals administered leptin. (From G. Legradi, C.H. Emerson, R.S. Ahima, J.S. Flier, R.M. Lechan, Leptin Prevents Fasting-Induced Suppression of Prothyrotropin-Releasing Hormone Messenger Ribonucleic Acid in Neurons of the Hypothalamic Paraventricular Nucleus, 1997, Endocrinology 138: 2569-2576.)

Circulating thyroid hormone levels also fall in association with severe illnesses and infection (287), but use a different set of regulatory controls.  This is based on the observation that both POMC and CART gene expression are increased in the arcuate nucleus (288) and circulating levels of leptin are elevated under these conditions (289).  In addition, norepinephrine secretion is increased in the PVN, and ordinarily would be expected to stimulate the secretion of TRH (290).  The precise anatomical pathways and mediators that override the activating effects of catecholamines, leptin and α-MSH on TRH neurons are not yet known.  However, type 2 iodothyronine deiodinase (D2), an enzyme that converts thyroxine into the more biologically active thyroid hormone, tri-iodothyronine, is expressed in tanycytes and D2 expression and enzymatic activity is substantially increased by endotoxin (291).   Given the location of tanycytes in the median eminence in contact with both the CSF and blood in the portal vascular plexus, and evidence that they express the thyroid hormone transporter, monocarboxylate transporter 8 (MCT8) (292), it is conceivable that tanycytes contribute to the fall in circulating thyroid hormone associated with infection by increasing the concentration of T3 in the mediobasal hypothalamus and suppressing the synthesis of TRH in hypophysiotropic neurons by local feedback regulation.  Hence, tanycytes may extract T4 from the bloodstream or the CSF, convert T4 to T3, and then release T3 into the CSF that could diffuse into the PVN by volume transmission (293)after moving between ependymal cells lining the third ventricle; release T3 directly into the median eminence that could be taken up by TRH axon terminals and then transported retrogradely to the PVN; and/or concentrate in arcuate nucleus neurons that have known projections to TRH neurons in the PVN (278, 282, 283, 294).  This mechanism is supported by observations demonstrating that the administration of LPS to D2 KO mice prevents the anticipated reduction in TRH mRNA observed in WT animals (295).  T3 may also be released into the portal capillary system for conveyance to the anterior pituitary and contribute to the mechanism whereby endotoxin inhibits the secretion of TSH.  The hypothesized regulatory mechanism is schematized in Fig. 34.

Fig. 34. Proposed mechanism for D2-regulation of the hypothalamic-pituitary-thyroid axis following the administration of LPS. LPS increases D2 activity in tanycytes resulting in increased T4 to T3 conversion. [1] T3 is released from tanycyte apical processes into the CSF for conveyance to the paraventricular nucleus, or [2] taken up from hypophysiotropic TRH axonal processes in the median eminence and transported retrogradely back to its cell body in the paraventricular nucleus. [3] T3 may also be released into the portal capillary system and directly inhibit the secretion of TSH. Local tissue hyperthyroidism inhibits TRH in the paraventricular nucleus. (From Lechan, R. M.; Fekete, C., Role of thyroid hormone deiodination in the hypothalamus. Thyroid 2005, 15, (8), 883-997.)

Cold exposure is another example of how afferent input to hypophysiotropic TRH neurons can alter the sensitivity of feedback regulation by circulating levels of thyroid hormone.  In this instance, however, TRH secretion is increased (296).  The mechanism involves catecholamine projection pathways from the brainstem to hypophysiotropic TRH neurons cell bodies and their axon terminals in the median eminence to increase both TRH mRNA and TRH release, respectively. 

 

REGULATION OF GnRH SECRETION

 

Pulses of GnRH initiate the pulsatile release of anterior pituitary gonadotropins, and changes in the GnRH pulse frequency dictate how much LH and FSH ultimately will be released (297,298).  This intermittent signal is of critical importance for pubertal development and necessary for the regulation and maintenance of normal reproductive function throughout the ovulatory cycle.  In the absence of episodic GnRH release, such as with continuous, exogenous administration of GnRH, the synthesis and secretion of gonadotropins are profoundly suppressed as a result of desensitization of GnRH receptors (299).  The central mechanisms governing the pulsatile secretion of GnRH may involve a variety of factors, but the most important would appear to be the recently discovered kisspeptin/G protein-coupled receptor 54 (GPR54) neuroregulatory system (300,301).  The kisspeptins derive from a single precursor but comprise a group of peptide molecules ranging from 10-54 amino acids, all capable of binding and activating the G-protein coupled receptor GPR54 with similar efficacy (301,302).  In humans, kisspeptin-54 has also been termed metastin on the basis that it was originally recognized to suppress cancer metastasis (303).

 

GnRH-producing neurons are located primarily in the preoptic region in rodents, but in all animal species give rise to axons that project caudally to terminate in the external zone of the median eminence (304).  Although GnRH neurons may have intrinsic oscillatory characteristics that might explain the pulsatile secretion of GnRH (2305.306), evidence would support the importance of GPR54 and the kisspeptins in modulating their secretory responses and in particular, the resurgence of GnRH pulsatility during puberty.  Namely, both humans and animals with GRP54 deficiency have hypogonadotropic hypogonadism despite normal development of GnRH neurons and normal LH and FSH secretion in response to GnRH (297,307), GPR54 mRNA is expressed by GnRH neurons (307), central and peripheral administration of kisspeptins potently induce gonadotropin secretion ( 308, 309), kisspeptin-induced LH secretion can be blocked with GnRH receptor antagonists (309,310), kisspeptin depolarizes ~90% of GnRH neurons in adult mice (311), and transgenic mice with targeted disruption of the Kiss1 gene (that gives rise to the kisspeptins) has an identical phenotype as transgenic mice deficient in GPR54 (312).  As brief intravenous infusions of kisspeptin every hour for 48h induce pulsatile LH discharges similar to those observed during puberty, whereas continuous kisspeptin infusion in mice or monkeys downregulate LH secretion by desentensitizing GRP54 (313-316), it has been proposed kisspeptin-producing neurons comprise the pulse generator for GnRH neurons or at the very least, amplify the activity of the pulse generator (317).

 

Neurons producing the kisspeptins are located in the hypothalamic arcuate nucleus and in some species, the anteroventral paraventricular nucleus (AVPV) (316), and express the alpha estrogen receptor (318).  Curiously, these two populations are regulated inversely to each other.  Thus, kisspeptin gene expression in arcuate nucleus neurons increases following ovariectomy and decreases with estrogen administration, whereas the reverse occurs in anteroventral paraventricular cells (315,316).   It has been proposed, therefore, that these two, kisspeptin neuronal populations may mediate the negative and positive feedback effects of estrogen on GnRH neurons (319), with the AVPV neurons involved in the estradiol/progesterone-induced preovulatory GnRH/LH surge (320).  Functional heterogeneity among the two kisspeptin neuronal populations is exemplified by the observation that kisspeptin neurons in the AVPV are sexually dimorphic, being particularly prominent in females, but virtually absent in males secondary to increased testosterone levels in males during development, and explain the inability of male rodents to mount a LH surge (321,322).  In addition, the arcuate nucleus population of kisspeptin neurons co-express neurokinin B and dynorphin (323), that contribute to the pulsatile release of GnRH through opposing actions on kisspeptin, neurokinin B stimulating and dynorphin inhibiting its secretion (324,325).  For this reason, the population of kisspeptin neurons is commonly referred to as KNDy neurons.  Phoenixin-20 amide (PNX), is also synthesized in kisspeptin neurons and may activate these neurons though an autocrine mechanism by binding to GPR173 receptors or through connections with other kisspeptin neurons (326).  Arcuate nucleus kisspeptin neurons also mediate the inhibitory effects of anorexia on reproductive function via a leptin-dependent mechanism (327).

 

Kisspeptin-containing axon terminals have been observed to terminate on hypophysiotropic GnRH neurons, but only the minority of cells and with surprisingly few boutons (315,317).  In contrast, kisspeptin-containing axons heavily inundate the median eminence and extensively intermingle with GnRH-containing axon terminals (315,317).  Although axo-axonal interactions between kisspeptin- and GnRH-containing axon terminals have not been observed (315), these specializations may not be required for physiologic function in the median eminence.  Evidence that peripheral administration of kisspeptin has a similar potent action on LH secretion as central administration (297), and physiologic data showing that exogenous administration of kisspeptin to hypothalamic explants deficient in GnRH neurons still potently release LH into the medium (313), strengthen the possibility that kisspeptin‘s actions may be directly on GnRH axon terminals in the median eminence.  Figure 35 summarizes a hypothesized mechanism for the neuroendocrine regulation of GnRH neurons by kisspeptin.

Figure 35. Simplified schematic demonstrating the mechanism for the regulation of GnRH secretion by kisspeptin. Two populations of kisspeptin neurons, oppositely regulated by estrogen, impinge on GnRH neurons in the medial preoptic nucleus that project to the neural-hemal contact zone in the median eminence. Kisspeptin-containing axon terminals may also interact with GnRH axon terminals in the median eminence.

Glial-neuronal interactions in the median eminence and in contact with GnRH cell bodies may also be involved in regulating the delivery of GnRH to the portal system. In the median eminence, two mechanisms have been proposed (328-331). The first involves the release of glutamate, prostaglandins and growth factors from tanycytes that induce the secretion of GnRH from their axon terminals in the median eminence. The second involves plastic rearrangements between tanycyte end foot processes and GnRH axon terminals, allowing or disallowing secreted GnRH from entering portal capillaries. Thus, it is proposed during the preovulatory gonadotrophin surge, estrogen binds to alpha type estrogen receptors on tanycytes and induces tanycyte end feet retraction through PGE2-dependent production of TGFβ1, allowing GnRH axon terminals to establish better contact with the portal vessels (332). Estrogen may also affect the synthesis of adhesion molecules such as polysialylated neuronal cell adhesion molecule (PSA-N-CAM) and synaptic cell adhesion molecule (SynCAM) that facilitate glial-neuronal interactions and remodeling (328,329). The release of nitric oxide from portal vessel endothelial cells may also participate in tanycyte retraction by affecting actin cytoskeleton remodeling (332). Astrocytes have also been observed to enwrap GnRH perikarya and may similarly induce the release of GnRH through the release of PGE2 and/or TGFβ (331). It has also been hypothesized that astrocytes contribute to the mechanism whereby GnRH neurons are synchronized to generate the pulsatile release of the neuropeptide (331).
Mechanisms for reawakening of the reproductive axis during puberty is not fully understood, but appears to involve a decrease in transsynaptic inhibition to the GnRH neuronal system and increase in its stimulatory input from afferent neurons. GABAergic neurons are one of the major inhibitory inputs to the GnRH system and when inhibited, result in premature activation of the GnRH neuronal system. Increased stimulatory drive has been associated with glutamatergic transmission as well as increases in norepinephrine and NPY. Circulating levels of leptin also have an important role in the central modulation of puberty and reproductive function by food availability and nutritional status and can reverse the suppressive effects of undernutrition on the reproductive axis, but mediated primarily through kisspeptin neurons. Conversely, inhibition of reproductive function in association with stress appears to be mediated through CRH as suppression of gonadotropin secretion is reversible by administration of a CRH antagonist (333).
MODULATION OF PROLACTIN-REGULATING FACTORS
Prolactin secretion from the anterior pituitary is primarily under inhibitory regulation by dopamine neurons (A12 group of Dahlstrom and Fuxe) located in the arcuate nucleus. However, a number of prolactin-releasing factors have also been identified including TRH, oxytocin, VIP, vasopressin, histidine isoleucine, and serotonin, that can also trigger prolactin release under different physiologic circumstances following direct release into the portal system (334). For example, histidine isoleucine, which is co-expressed with CRH in parvocellular neurons in the PVN, and vasopressin are involved in stress-induced prolactin secretion through direct effects on lactotrophs. Serotonin has been implicated in lactation-induced prolactin secretion partly by stimulating TRH secretion from hypophysiotropic neurons, but also by inhibiting dopamine secretion and is further discussed below in the Lactation section.
Feedback control of prolactin secretion is mediated by a short-loop mechanism in which prolactin, itself, increases dopamine synthesis from the A12 neurons However, multiple neurotransmitter systems impinge on the A12 neurons that contribute to regulation of their neurosecretion including cholinergic neurons derived from the basal forebrain, hypothalamic glutamatergic and brainstem serotoninergic afferents that have activating effects and hypothalamic histaminergic and opiate peptide-containing afferents that have inhibitory effects (334).
Modulation of Vasopressin Secretion and Osmoregulation
Maintenance of the appropriate solute concentration in plasma (osmotic homeostasis) and plasma volume (volume homeostasis) is dependent upon two major factors, the perception of thirst and the ability to synthesize and secrete the antidiuretic hormone, arginine vasopressin from magnocellular neurons in the hypothalamic PVN (335). These two factors are closely interrelated such that amount of vasopressin circulating in the periphery is proportional to the plasma osmolality (336). Vasopressin induces cAMP and the translocation of specific aquaporin-2 water channels to the apical plasma membrane of tubular epithelial cells in the kidney, allowing water resorption (337,338). In addition, the rise in osmolality has independent behavioral effects. Thus, when plasma osmolality rises above basal levels, there is inducement to drink, shortly following the rise in vasopressin (335). While some vasopressin neurons in the PVN are intrinsically osmosensitive (339,340), the major mechanism of osmoregulation is via afferent pathways originating from osmoreceptor cells in other neuronal populations. These include inputs from the OVLT and the median preoptic nucleus, which if damaged, simultaneously abolish vasopressin secretion and thirst responses to hyperosmolality in both experimental animals and man (136,341). The SFO is also activated by a rise in osmolality and may contribute to vasopressin release through direct afferent projections to the PVN and/or to the OVLT using angiotensin II as a mediator (133-136,342). As mice with targeted disruption of the transient receptor potential (TRP) ion channels, TRPV1 and TRPV4, have impairment in vasopressin secretion and reduced drinking in response to hypertonic stimuli and show diminished cFos responses in the OVLT, these ion channels may be responsible for osmoreception (343). Along these lines, it particularly interesting that vasopressin can also be increased by hyperthermia (344,345), and that the trp genes are known to encode proteins involved in thermoregulation (see section E. Thermoregulation). Indeed, TRPV1 is required for thermosensory transduction of vasopressin secretion from isolated magnocellular neurons (346). Sodium channels may also contribute to the regulation of vasopressin secretion in response to hypernatremia by acting on neurons in the OVLT and SFO (347).
Whereas forebrain pathways communicate information about osmolality to the PVN, brainstem projections tend to carry nonosmotic, baroregulatory information, and important for vasopressin secretion, particularly in association with hypovolemia and hypotension (136,335). This information is carried through the vagus and glossopharyngeal nerves to the NTS and ventral lateral medulla, and then to the PVN through the ascending catecholaminergic pathways (Fig. 36). Magnocellular neurons in the PVN appear to be primarily innervated by the A1 catecholamine-producing cells in the ventral lateral medulla (247).

As described for hypothalamic tuberoinfundibular neurons, the threshold for vasopressin secretion by neurons of the magnocellular neurosecretory system can also be modified by their afferent signals as well as circulating factors.  For example, the osmotic threshold for vasopressin release can be altered by glucocorticoids.  Dexamethasone attenuates the vasopressin response to salt loading (348) and hypoadrenalism is commonly associated with the syndrome of inappropriate secretion of antidiuretic hormone (SIADH) that can be corrected by glucocorticoid administration (349).  These effects are exerted directly on vasopressin neurons given the presence of glucocorticoid receptors in these cells (350).  Other causes for SIADH, however, such as pulmonary disease and central nervous system disorders may be mediated through afferent pathways to the PVN.  Hormone mediators of these projections include vasoactive intestinal polypeptide (VIP), acetylcholine, angiotensin II, neuropeptide Y and noradrenaline, among numerous others (351,352).

 

Appetite and Satiety

 

The demonstration that discrete regions of the hypothalamus control food intake was based on the early studies of Hetherington and Ranson (353) in 1940 showing that localized lesions of the hypothalamus result in obesity.  These observations were seemingly confirmed in man when Reeves and Plum (354) reported that a discrete lesion (hypothalamic hamartoma) involving the hypothalamic ventromedial nucleus in a 28-year-old woman was associated with increased food consumption and profound obesity.  In contrast, bilateral lesions of the lateral hypothalamus in animals result in anorexia and weight loss (353). Thus, the concept of a hypothalamic ventromedial nucleus satiety center and lateral hypothalamic orexigenic center that can be influenced by peripheral signals was developed, and dominated thinking about the hypothalamic control of feeding for several decades.

 

It was not until 1994, however, when the discovery of leptin revolutionized thinking on the mechanisms governing appetite and satiety (355).  Leptin serves as an important humeral signal that reflects body fat stores, and by acting on discrete regions in the hypothalamus, orchestrates the behavioral, metabolic, and neuroendocrine adaptations to nutrient availability (285,286,356-359).  Thus, during nutrient abundance, leptin secretion is increased, leading to decreased appetite and increased caloric disposal, whereas nutrient insufficiency leads to decreased leptin secretion, resulting in increased appetite, energy conservation, and a shift to a neuroendocrine profile that facilitates metabolic adaptation.

 

A major site of leptin’s action is the mediobasal hypothalamus, primarily the hypothalamic arcuate nucleus via specific receptors (Ob-Rb) that influence the activities of two separate groups of neurons with opposing functions through a number of signaling pathways that include Jak-STAT, PI3k-Akt-FoxO1, SHP2-ERK, AMPK, mTOR-S6K (360-362).  These neurons include α-MSH-producing neurons that co-express CART, and AGRP neurons that co-express NPY (317).  These neurons send monosynaptic projections to identical target regions within discrete regions of the hypothalamus where the signals are integrated and then relayed by independent pathways to regions of the brain governing feeding behavior, energy expenditure, and hypophysiotropic function (286,356-358).  When circulating leptin levels are suppressed, such as during fasting, expression of genes encoding proteins that promote weight loss, and energy expenditure, α-MSH and CART, are reduced simultaneously with a marked increase in the genes encoding proteins that promote weight gain and reduce energy expenditure, AGRP and NPY.  Cooperation between the opposing components of the regulatory system governing appetite and satiety is underscored by the biological action of AGRP as both a competitive antagonist and inverse agonist at melanocortin receptors (MC3r and MC4r) (285,363).  Thus, during fasting, the rise in AGRP cooperates in down regulation of melanocortin signaling by antagonizing the action of α-MSH concurrently with inhibition of the POMC gene.  Reciprocal connections between the arcuate nucleus NPY/AGRP neurons and α-MSH/CART neurons also are present (364), suggesting an even greater complexity to this regulatory system.

 

It is becoming increasingly recognized that the melanocortin signaling system may be the predominant regulatory system governing appetite and satiety.  Whereas animals with targeted deficiency of NPY have an essentially normal phenotype and intact responses to fasting (365), animal models with targeted deletion of the type 4 melanocortin receptor (MC4r) and in humans bearing mutations that interfere with the function of the MC4r, the POMC gene, or the processing enzymes necessary to generate a fully mature α-MSH, develop a severe obesity syndrome (366-369).  Loss of tone in the melanocortin signaling system as a result of senescence of the arcuate nucleus POMC neurons may also explain the tendency for weight gain with aging (370,371).  Conversely, studies by Wisse et al (372) and Marks et al (373) have demonstrated that cancer cachexia can be prevented in experimental animals by the administration of melanocortin receptor antagonists.  Maintaining adequate tone in the melanocortin signaling system, therefore, would appear to have an especially important role in the maintenance of normal body weight.  Taking advantage of this observation is evidence that the MCRr agonist setmelanotide, has been shown to promote weight loss in individuals with leptin receptor or POMC deficiency (3740.

 

The arcuate nucleus is a main sensor of circulating levels of leptin, and by projecting to several different regions in the brain, provide the mechanism whereby leptin is capable of integrating a host of responses involved in energy homeostasis.  The arcuate-PVN projection pathway has an important role in regulation of the thyroid axis (see above).  Thus, hypophysiotropic TRH neurons in the medial parvocellular PVN receive direct projections from NPY/AGRP- and α-MSH/CART-producing neurons in the arcuate nucleus, altering the set point at which circulating thyroid hormone inhibits TRH (158,243,244,247) The end result during fasting is suppression of the HPT axis as a way to reduce energy expenditure.  Other targets in the PVN include neurons in the anterior and ventral parvocellular subdivisions of the PVN on the basis that these neurons show phosphorylation of CREB following the central administration of α-MSH (375).  Both subdivisions receive a high density of axons containing α-MSH and AGRP derived from the arcuate nucleus (376,377).  Thus, these regions may be involved in some of the other actions of leptin including the regulation of feeding and/or energy disposal.   This concept is supported by the observation that focal injections of α-MSH or α-MSH agonists directly into the PVN reduces feeding and can fully replicate the reduced feeding responses following icv administration (378).  Conversely, α-MSH antagonists injected into the PVN have a potent effect to increase feeding (379).  Since anterior parvocellular PVN neurons project to the limbic system (lateral septum and amygdala) (380,381), it is possible that this part of the PVN is involved in the behavioral manifestations of feeding.  The ventral parvocellular subdivision is involved in the regulation of the autonomic nervous system through descending projections to brainstem and spinal cord targets (164,381,382).  This region, therefore, may be involved in the regulation of energy disposal by controlling heat loss from brown adipose tissue through effects on uncoupling protein-1 (UCP-1) (383) and by affecting lipolysis and proteolysis in white fat and muscle, respectively (384).  A population of POMC-responsive, glutamatergic PVN neurons descends to the parabrachial nucleus, now also considered to be an important brainstem nucleus involved in the suppression of appetite (385).  Some parabrachial neurons project to the central nucleus of the amygdala using calcitonin gene-related transcript as a transmitter (386), implicating the central nucleus of the amygdala as another important node in the satiety pathway.  Descending projections from leptin-responsive PVN neurons may also modulate the sensitivity of brainstem neurons to satiety signals originating from the gastrointestinal tract, mediated at the level of brainstem nuclei (387,388), and have been shown to affect the anorexigenic effects of peripherally administered CCK (389).  Oxytocin may be one of several PVN-derived peptides relaying forebrain information about appetite and satiety to the brainstem (390).

 

In addition to the PVN, leptin-responsive neurons in the arcuate nucleus synapse on two separate populations of neurons in the lateral hypothalamus that produce melanin-concentrating hormone (MCH) and orexin (391).  These neurons project to multiple regions of the brain including the cerebral cortex, midbrain and pons including the ventral tegmental area (VTA) and the nucleus accumbens, well-recognized reward centers involved in hedonic eating and addiction (392).  MCH acts as an endogenous stimulator of food intake and its mRNA is increased during fasting (393,394) whereas orexin promotes arousal responses (395) that would have an essential role in permitting food-seeking behavior during periods of nutrient deficiency.  Orexin also increases gastric contractility through projections to the brainstem, and by reducing gastric distension, suppresses satiety signals carried by the vagus nerve (396).  Thus, during caloric restriction, activation of orexin- and MCH-producing neurons would have several actions to promote increased food ingestion and promote weight gain through effects on appetite, behavior and the incentive to feed.  Leptin-responsive, CART-producing neurons in the arcuate nucleus also project directly to the intermediolateral cell column of the spinal cord (397), indicating their importance in autonomic control.

 

Leptin receptors are also expressed by other hypothalamic nuclear groups including the caudal part of the hypothalamic dorsomedial nucleus (DMN) and the dorsomedial division of the ventromedial nucleus (VMN) (398).  Evidence that leptin-responsive regions of the brain other than the arcuate nucleus contribute to the regulation of feeding has been demonstrated by only a modest reduction in hyperphagia after the leptin receptor has been re-expressed selectively in arcuate nucleus of animals with leptin deficiency (399,400).  The DMN has extensive projections to the PVN, particularly portions involved in autonomic control, as well as direct brainstem projections to the dorsomotor complex of the vagus (401).  In addition, the DMN receives extensive projections from the arcuate nucleus, including projections from NPY/AGRP- and α-MSH/CART-leptin-responsive neurons (282,283).  As lesions of the DMN produce hypophagia and reduce linear growth, this nucleus has an important role in the homeostatic control of feeding behavior (402).  The DMN may also mediate the anorexic effect of cholecystokinin-8 (CCK-8) (403) through projections from CCK-8-producing neurons in the superior lateral subdivision of the parabrachial nucleus (404).  The VMN has been long implicated in the regulation of feeding behavior as lesions of the VMN produce hyperphagia (405).  However, these observations were likely due to transection of surrounding fiber pathways.  Because the dorsomedial portion of the VMN projects to the subparaventricular zone, it has been proposed that leptin-sensitive VMN neurons may have a role in circadian rhythms (286).

 

The brainstem also contributes to leptin-regulated neuroendocrine responses involved in feeding, particularly the dorsal vagal complex (DVC) (406).  The DVC is an important relay center in the brainstem for visceral sensory information carried by the vagus nerve from the liver GI tract, but also receives descending information from the forebrain including the PVN (356).  Glucagon-like peptide (GLP-1)-producing neurons in the DVC have been shown to be leptin responsive (407), and this peptide has potent anorexic effects (408).  NPY-producing neurons in the hypothalamic arcuate nucleus have been proposed as the primary target for GLP-1 due to the high concentration of GLP-1 receptors in this region and because GLP-1 can inhibit NPY-induced feeding (409,410).  However, the arcuate nucleus contains relatively few GLP-1-containing nerve terminals as compared to the PVN and DMN (411), and discrete injections of GLP-1 into the PVN are capable of suppressing feeding (412).  Leptin also enhances the anorexic effects of CCK (see below) that can be diminished by selective knock-out of leptin receptors in the DVC (413). Leptin also has important effects on dopaminergic neurons in the ventral tegmental area (VTA), that mediates motivated and reward-seeking behaviors including food consumption, as selective knock-out of the leptin receptor in this area results in increased feeding (414).

 

In addition to leptin, a number of gut peptides contribute to the central regulation of appetite and satiety (Table 10), either by acting directly on the hypothalamic arcuate nucleus or transmitted through the DVC via vagal and spinal afferent neurons (415-417).  Among these are insulin, cholecystokinin (CCK), peptide YY (3-36), pancreatic polypeptide (PP), GLP-1, amylin, oxyntomodulin and ghrelin (418).  Like leptin, levels of insulin vary with adiposity (316) and are suppressed by fasting and increased by eating.  In addition, the intracerebroventricular administration of insulin reduces food intake and body weight (419) and prevents fasting-induced increases of NPY and AGRP mRNA in arcuate nucleus neurons (420).  Amylin is co-released with insulin in response to a meal and functions as an anorectic hormone through effects on serotonin, dopamine and histamine (421).  Both CCK and pYY are secreted by the gut in response to feeding and may have a role in the termination of eating, CCK through effects on receptors in visceral sensory axons that travel in the vagus nerve (422) and pYY through blood-borne inhibitory signals exerted through Y2 receptors on NPY/AGRP neurons in the hypothalamic arcuate nucleus (423).  In addition to the central GLP-1 system described above, GLP-1 is also released into the circulation after a meal from the distal small intestine and colon, decreasing food intake through a vagal mechanism (424).  Oxyntomodulin, which like GLP-1 derives from the same intestinal cells by posttranslational processing of proglucagon, binds to the GLP-1 receptor and has similar actions on reducing food intake that are abolished in the GLP-1 receptor knockout mouse (425).  However, this peptide also appears to increase energy expenditure, raising the possibility of its particular utility in the treatment of obesity (426).  As its name suggests, PP derives from the pancreas and inhibits food intake by binding to Y4 receptors in the DVC and arcuate nucleus and through effects mediated by the vagus nerve (427).  Ghrelin, produced primarily by the stomach, is secreted directly into the bloodstream, but as opposed to all other identified gut-derived peptides, ghrelin stimulates food intake and is highest just prior to a meal and falls after eating (428).  Thus, ghrelin may have a primary role in meal initiation.  Its target is also the hypothalamic arcuate nucleus, increasing the expression of NPY/AGRP mRNAs (429) and inhibiting POMC neurons (430).  However, vagotomy abolishes ghrelin-stimulated eating (431), indicating that ghrelin may also signal through the brainstem.  Hypersecretion of ghrelin has been proposed as a mechanism for the morbid obesity associated with the Prader Willi syndrome (432).  Nevertheless, treatment of Prader Willi patients with somatostatin analogues that suppresses circulating levels of ghrelin, does not improve the hyperphagia associated with this disorder (433), indicating that other mechanisms are responsible or that compensatory mechanisms take place.  A second peptide derived from the ghrelin gene has been recently identified, obestatin, but its role in the regulation of appetite requires further characterization.  Curiously, obestatin appears to suppress food intake (434).  Nesfatin-1 has also been found to co-localize with ghrelin in gastric cells (435) and has potent satiety effects (436).  While circulating nesfatin-1 can cross the blood-brain-barrier and inhibit NPY neurons in the arcuate nucleus (437), the peptide is highly expressed in the lateral hypothalamus in MCH neurons (438) and in the PVN and may be under the regulation of -MSH (436).  New and potentially exciting chapters in gut-brain signaling relates to the importance of bile acids (439) and gut microbiota (417) in influencing central neural signaling primarily to affect energy homeostasis, but the mechanisms by which this takes place have yet to be fully elucidated.

 

Thyroid hormone should probably also be considered as a circulating hormone involved in appetite regulation through actions on the hypothalamus.  Thyrotoxicosis is commonly associated with hyperphagia both in experimental animal models and humans (440-444), although the mechanism(s) by which this occurs are uncertain.  Ishii et al (442) have shown an increase in hypothalamic NPY mRNA following the administration of thyrotoxic doses of T3, perhaps secondary to T3-induced increase in neuronal uncoupling protein 2 in NPY neurons (445).  Nevertheless, T3-associated hyperphagia is only partially attenuated by a NPY receptor antagonist, indicating that other mechanisms must also be operable.  The hypothalamus is richly endowed with thyroid hormone receptors (446,447), and nuclear T3 concentrations in hypothalamic extracts are elevated following the systemic administration of thyrotoxic doses of T3 or T4 (448).  Recent studies by Kong et al (449) have demonstrated that the systemic administration of superphysiologic doses of T3 increase immediate early gene expression in the hypothalamic ventromedial nucleus.  In addition, microinjection of T3 into the hypothalamic ventromedial nucleus induces a 4-fold increase in food intake during the first hour following injection.  These data suggest a direct effect of T3 on the hypothalamus to induce feeding.  Evidence that the nuclear T3 concentrations in the hypothalamus are elevated following the systemic administration of thyrotoxic doses of T3 or T4 (448) further substantiates this hypothesis.  The coincidence that rats display a nocturnal pattern of feeding and that hypothalamic deiodinase activity follows a circadian pattern that increases during the night which may have the effect of increasing local tissue levels of T3 (444,449), provides circumstantial evidence for the potential importance of thyroid hormone to regulate feeding by increasing local, neural tissue concentrations of T3.

 

Estrogen also has an antiobesity effect and may contribute to the tendency for weight gain following menopause.  The effects of estrogen appear to be mediated by the estrogen receptor-alpha (ERα), as mice with targeted deletion of this gene develop obesity (450).  ERα is expressed by POMC neurons and when selectively ablated from these neurons, hyperphagia ensues (451).

 

Adipose-derived peptides (adipokines) other than leptin are also believed to have important roles in the regulation of appetite and energy expenditure (452).  In particular, adiponectin serves as a starvation signal to preserve fat stores, increasing in the plasma with fasting.  Adiponectin, thereby, functions inversely to leptin in the brain by binding to the adiponectin receptor 1 (AdipoR1) in the arcuate nucleus, and by activating AMPK, increases food intake and reduces energy expenditure by suppressing UCP-1 in brown adipose tissue (453).

 

Recent attention has also been given to the role of neurotransmitters and endocannabinoids in the regulation of food intake and energy expenditure (454,455).  The cannabinoid receptor, type 1 (CB1) is the primary cannabinoid receptor in the central nervous system (456), and has a widespread distribution that includes the hypothalamus and limbic structures (457).  Endogenous ligands for CB1, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG), inhibit synaptic release of transmitters in both excitatory and inhibitory terminals (458).  Since endocannabinoids are synthesized by neurons postsynaptic to axon terminals containing CB1 receptors, they are actually a retrograde signaling system.  Neuronal release of endocannabinoids, therefore, regulates the activity of the cells’ own CB1-containing innervation.  Endocannabinoids exert orexigenic effects when injected into the hypothalamus and may mediate the effects of ghrelin to increase food intake, block the inhibitory input to MCH neurons in the lateral hypothalamus and attenuate the excitatory input of anorexigenic neurons in the paraventricular nucleus (459,460).  Endocannabinoids also activate the mesolimbic dopamine system that is involved in hedonistic eating (see below).

 

With respect to neurotransmitters, selective removal of the GABA transporter from AGRP neurons, for example, results in a lean phenotype, even on a high fat diet (461), whereas the selective deletion of leptin receptors from all neurons that express the GABA vesicular transporter develop marked hyperphagia (462).  Serotonin is also known to have a profound effect on food intake, probably by acting on a subset of POMC neurons in the hypothalamic arcuate nucleus that express serotonin 2C receptors (463).  Thus, two populations of POMC neurons have been proposed, those responsive to leptin that increase energy expenditure and those responsive to serotonin that reduce food intake (462, 464).  These observations are in concert with the observation that serotonin agonists such as fenfluramine and lorcaserin have had efficacy in the treatment of obesity (465).

 

Heterogeneity of POMC neurons in the arcuate nucleus as defined by GABAergic and glutamatergic phenotypes has recently been recognized (466), although the physiologic significance is still uncertain.  As noted above, dopamine has also been associated with reward-related food intake mediated through leptin-responsive, dopamine-producing neurons in the VTA (467).  Dopamine has a major role in the regulation of reward processing including the desire for drugs of abuse and sweet foods (468).  Recent evidence has linked impaired dopamine transmission in the mesolimbic nervous system to dietary obesity (469).

 

Nutrient sensing by the brain also appears to contribute to the regulation of appetite and satiety.  Hypoglycemia increases food intake, presumably as a result of glucose deprivation on brainstem catecholamine neurons which project to the hypothalamus (470); the amino acid, L-leucine, but not other branched chain amino acids, exerts its anorexic actions by activating the mammalian target of rapamycin, mTORC1 pathway, in arcuate nucleus NPY/AGRP neurons by reducing NPY and AGRP  (471) and in the caudomedial nucleus of the NTS (472); and fatty acids inhibit food intake by suppressing AMP-activated protein kinase (AMPK) in arcuate nucleus which similarly reduces NPY (473).  Alpha-MSH-producing neurons in the arcuate nucleus are also excited by glucose-mediated closure of ATP-sensitive potassium (KATP) channels (474).  The importance of this mechanism has been shown by Parton et al (475), demonstrating that expression of a mutant form of the KATP channel subunit Kir6.2b in α-MSH-producing neurons that impairs ATP-mediated closure of KATP channels, results in impaired glucose tolerance.

 

In addition to the major role of the hypothalamus and brainstem in the regulation of eating, many other regions of the brain are also involved, particularly in the human brain (476).  As noted above, dopaminergic neurons in the ventral tegmental area and substantial nigra that project to the nucleus accumbens, striatum and orbitofrontal cortex are involved in hedonic feeding, leading individuals to seek highly rewarding, high caloric foods.  Emotional eating may be mediated by projections from the amygdala to the hypothalamus to initiate a hunter response.  Finally, cognitive control systems in the prefrontal cortex can allow for self-restraint, even though food may seem highly appealing.

 

 A summary diagram of the complex mechanisms involved in the regulation of appetite and satiety is shown in Fig. 37.

 

Figure 37. Simplified schematic representation of some of the regulatory factors and pathways involved in the regulation of appetite and satiety. Circulating factors derived from fat, pancreas, liver and the gastrointestinal tract converge on the hypothalamus and/or brainstem to orchestrate a series of responses that promote increased appetite, decreased energy expenditure, activation of mesolimbic reward centers and reduce circulating thyroid hormone levels. Note similarities of target regions in the brain by several regulatory factors, particularly for AGRP/NPY and -MSH/CART neurons in the hypothalamic arcuate nucleus. The endocannabinoid system exerts regulatory effects on neurons in multiple regions of the brain. Other regions of the brain including the amygdala, hippocampus, and prefrontal cortex also have important roles in the regulation of eating. ARC= arcuate nucleus, DMN= dorsomedial nucleus, DVC= dorsal vagal complex, LH= lateral hypothalamus, PVN= paraventricular nucleus, SNS=sympathetic nervous system, VMN= ventromedial nucleus.

Lactation

 

Suckling is a well-recognized physiological stimulus for increased prolactin secretion from the anterior pituitary gland and oxytocin from the magnocellular, neurohypophysial system (477).  This stimulus is relayed to the hypothalamus via the spinal cord and brainstem (478,479), although the precise intrahypothalamic pathways involved have not been precisely elucidated (480).  Using c-fos as a marker for neuronal activation, several potential relay centers in the brainstem have been identified including the ventrolateral medulla (A1 catecholamine cell group), locus coeruleus, lateral parabrachial nucleus, caudal portion of the paralemniscal nucleus, and lateral and ventrolateral portions of the caudal part of the periaqueductal gray (481).  By inhibiting tuberoinfundibular dopamine neurons in the arcuate nucleus and increasing prolactin releasing factors in tuberoinfundibular neurons in the hypothalamic PVN and posterior pituitary (482), these signals permit high circulating levels of prolactin, particularly when dopamine has been inhibited.  Putative prolactin releasing factors include thyrotropin-releasing hormone, oxytocin, serotonin, opioid peptides and TIP39, although the latter may function by increasing dynorphin (480).  Prolactin also feeds back on dopamine neurons in the arcuate nucleus to inhibit the release of dopamine (483), but this ultrashort feedback loop is inhibited during lactation by dissociation of electrical activity and dopamine release from dopamine neurons, allowing for sustained elevations in prolactin in response to suckling (484).  Prolactin is not only important for the synthesis and maintenance of milk secretion, but contributes to the physiologic responses that complement lactation including the development of material behavior and inhibition of reproductive function (477).  These effects are facilitated by upregulation of prolactin receptors in the choroid plexus during lactation (485), allowing increased entry of prolactin from the circulation into the brain through a carrier-mediated transport system, as well as in several hypothalamic nuclear groups including the medial preoptic nucleus, PVN, supraoptic nucleus and arcuate nucleus (486).  In particular, binding of prolactin to its receptor in the medial preoptic nucleus may be important for maternal behavior, as antagonizing prolactin receptors with a specific prolactin receptor antagonist injected directly into the preoptic area delays the onset of material behavior (487).  In addition, inhibition of pulsatile gonadotropin secretion associated with lactation may be at least partly mediated by the effects of prolactin on GnRH neurons in this region (477), although recent evidence for suckling-induced inhibition of kisspeptin gene expression in arcuate nucleus neurons may also be contributory (488.489).

 

Suckling also results in a marked increase in NPY gene expression in arcuate nucleus neurons (490,491), mediated both by neuronal afferents relayed from the brainstem and by the fall in circulating levels of leptin brought about by negative energy balance resulting from milk production (492).  As described above (see C. Appetite and Satiety), NPY is a powerful orexigenic substance.  Its increase during suckling, therefore, has been proposed to explain the hyperphagia associated with lactation to meet the energy demands imposed by milk production (492), although other changes are also observed in arcuate nucleus neurons that may contribute to hyperphagic responses including an increase in AGRP gene expression and decrease in POMC and CART mRNAs (493,494), similar to that described above for fasting.  NPY-containing axon terminals of arcuate nucleus origin also richly innervate the medial preoptic region and can be found in close proximity to GnRH neurons (495).  The suckling-induced rise in NPY, therefore, may also contribute to inhibition of reproductive function (494) through direct actions on GnRH neurons by binding to NPY Y5 receptors (496).

 

The other major component of the suckling response is the effect on oxytocin release from the posterior pituitary to allow milk ejection.  While some of the anatomical pathways responsible for this response may parallel with those inducing prolactin secretion, distinct pathways are also likely to exist.  Particularly remarkable about this mechanism is that during suckling, oxytocin neurons on both sides of the hypothalamus in the paraventricular and supraoptic nucleus are synchronized to release oxytocin in a pulsatile manner throughout the suckling stimulus.  This coordinated pattern of release may be regulated by afferent inputs to the oxytocin neurons that include glutamatergic neurons arising in the lateral septum and bed nucleus of the stria terminalis (480).  Anatomic plasticity of the oxytocin neurosecretory system also contributes through reraction of astoglial processes that normally separate oxytocin neurons, allowing for increased somatic appositions (497).  Local release of oxytocin from somata and dendrites (intranuclear release) in response to suckling may also contribute by modulating the effect of neurotransmitters, increasing oxytocin gene expression and through local effects on astroglia to promote somatic appositions (480).

 

The circuitry involved in the coordinated responses to suckling is illustrated in Fig. 38.

 

Figure 38. Schematic drawing of the major pathways involved in lactation. Suckling leads to neurogenic responses mediated through the medulla to inhibit dopamine secretion in arcuate nucleus (ARC) neurons and stimulate oxytocin (OXY) secretion in paraventricular (PVN) neurons. Prolactin and oxytocin exert effects on the breast, but prolactin also gains entry into the CNS to affect maternal behavior and inhibit reproductive function by acting on medial preoptic neurons (mPOA), and further inhibit the secretion of dopamine from ARC neurons. Milk production leads to a fall in circulating levels of leptin causing an increase in NPY and AGRP and inhibition of POMC in ARC neurons. NPY is also increased by neurogenic signals from the brainstem. Inhibitory effects of NPY on GnRH-producing neurons in the mPOA contributes to inhibition of reproductive function. NPY also exerts direct effects on the PVN to induce increase feeding and promote energy conservation.

 

Thermoregulation

 

The hypothalamus is the primary locus for coordinating thermoregulatory information and integrating thermoregulatory responses (498-500).  It continually monitors local brain temperature through temperature sensitive neurons and by utilizing thermoreceptors in the skin, abdominal cavity and spinal cord, and then orchestrates a series of responses to maintain normal, core body temperature by utilizing the autonomic nervous system, altering behavior, and through neuroendocrine responses.  Thyroid hormone is a necessary component for heat regulation since in its absence (myxedema), hypothermia commonly develops.

 

Although thermosensitive neurons can be found throughout the hypothalamus (401), the most important thermoregulatory locus is the preoptic region including neurons in medial and lateral portions of the preoptic nucleus, anterior hypothalamus including the perifornical region, dorsomedial nucleus and nearby regions of the septum.  Although most of the data on the anatomy of thermogenesis arises from work in rodents, evidence that the preoptic/anterior hypothamus is also the critical hypothalamic area for regulating thermogenesis in humans is apparent by functional MRI (502).  Preoptic cooling increases heat production by inducing shivering, or by nonshivering thermogenesis mediated by sympathetic activation of uncoupling protein-1 (UCP-1) in brown adipose tissue that allows mitochondria to generate heat from ATP, and by increasing intermediary metabolism in muscle and other parenchymal organs.  In addition, cooling induces heat retention responses by cutaneous vasoconstriction and redirecting blood flow from cutaneous to deep vascular beds, results in behavioral responses (seek warmer environment, put on more cloths, increase food intake), and in some animal species, increases thyroid thermogenesis by activating the hypothalamic-pituitary-thyroid axis (503).  Conversely, preoptic warming reduces heat production and increases heat loss responses through vasoconstriction, sweating, increased respiration (panting), inhibition of UCP-1 in brown adipose tissue, and specific behavioral responses (501).

 

The recent discovery of substantial amounts of brown adipose tissue are also present in adult humans (504,505) has indicated that like smaller mammals, brown adipose tissue has a similar role in whole body thermogenesis in man.  Just a few grams of brown adipose tissue may be adequate to increase daily energy expenditure in humans by ~20% (506).  Indeed, knowledge of the regulatory circuits and factors that govern thermogenesis in brown adipose tissue (see above section on Appetite and Satiety) has provided insight for the development of innovative molecular and pharmacological approaches to achieve weight loss (507), although still at a preliminary stage.

 

Two types of thermosensitive neurons can be found in the preoptic region, warm sensitive neurons that increase their firing rate when preoptic temperature rises and cold sensitive neurons that increase their firing when preoptic temperature falls (501).  However, the warm sensitive neurons predominate both in cell number and in importance of the regulatory responses for both heat loss and heat production mechanisms.  Thus, lesions involving the preoptic region per se are commonly characterized by abnormalities in heat dissipation and lead to hyperthermia and elevated temperature in brown adipose tissue (502).  Surprisingly, the neurotransmitter/peptide mediators and pathways mediating thermoregulatory responses are not precisely known.  When injected directly into the preoptic area, however, a number of different substances can induce hypothermic or hyperthermic responses (Table 11).  Since thermoregulation involves the coordination of multiple responses that can differ between animal species (i.e., panting in the dog, increased salivation in the rat which can be applied to the fur to enhance evaporative heat loss, sweating in man), it is logical that several different pathways are utilized.  Evidence suggests that efferent pathways governing shivering involve ipsilateral and crossed fibers (508) that traverse the median forebrain bundle to terminate in the posterior hypothalamus, using GABA as a neurotransmitter (509).  The pathway continues caudally through the midbrain, dorsolateral to the red nucleus, and interacts with reticulospinal neurons.  It then proceeds through the reticulospinal tract, to innervate α-motor neurons in the ventral horn of the spinal cord.  Regulation of heat production in brown adipose tissue also proceeds from preoptic neurons through the medial forebrain bundle to hypothalamic nuclear groups involved in autonomic regulation, particularly the PVN, FiDMN and VMN (509).  The PVN has direct efferent projections to preganglionic neurons in the intermediolateral column of the spinal cord which give rise to the sympathetic innervation of brown adipose tissue (510).  Regulation of cutaneous blood flow also proceeds from thermosensitive neurons in the preoptic region through axons descending in the medial forebrain bundle, but likely relayed to neurons in ventrolateral portions of the midbrain periaqueductal gray (PAG) before proceeding to sympathetic preganglionic neurons in the spinal cord.  PAG neurons show strong c-fos induction following unilateral preoptic region heating (511) and induce cutaneous vasodilatation when stimulated (512).  Preoptic warming also inhibits vasoconstrictor neurons in the medullary raphe (raphe magnus and pallidus) (513) that have projections directly to preganglionic sympathetic neurons in the intermediolateral column of the spinal cord (514).   Pathways mediating behavioral changes associated with thermoregulation are unknown.

 

Table 11.  Substances That Exert Thermoregulatory Effects in the CNS

HYPOTHERMIC

HYPERTHERMIC

ACETYLCHOLINE                                        

ANGIOTENSIN II                                           

CCK                                                               

DOPAMINE                                                    

ESTROGEN                                                    

α-MSH                                                            

NEUROTENSIN                                             

NOREPINEPHRINE

OPIOID PEPTIDES

SOMATOSTATIN

SUBSTANCE P

VASOPRESSIN

CRH

GABA

OPIOID PEPTIDES

PROGESTERONE

PROSTAGLANDINS

SEROTONIN

TRH

 

 

While it is clear that the transient receptor potential ion channels, including TRP vanilloid 1 (TRPV1), TRPV3, TRPV4, and TRPV8, are involved as heat receptors in the periphery (515), it remains controversial whether these receptors contribute to thermoregulatory responses at the level of the preoptic nucleus (516).  TRPV1 mRNA is present in the hypothalamus, and administration of capsaicin, a ligand for TRPV1, directly into the preoptic hypothalamus induces hypothermic responses, suggesting activation of warm-sensitive neurons (517,518).  Nevertheless, mice deficient in TRPV1 show normal thermoregulation when placed in a warm environment (519), raising questions about the physiologic importance of TRPV1 in central thermoregulatory responses.   Recently, however, melastatin type 2 ion channels (TRPM2) have been identified in the preoptic nucleus that when selectively activated or inhibited results in profound hypothermia or hyperthermia, respectively (520).  In addition, transgenic animals deficient in TRPM2 have an exaggerated fever response.

 

Under normal circumstances, there is a diurnal variation of body temperature, highest in late afternoon and early evening and lowest in morning upon arising.  The hypothalamic suprachiasmatic nucleus controls this rhythm, and would appear to do so through direct projections to the dorsal portion of the subparaventricular zone, a region just ventral to the PVN (see F. Circadian Rhythmicity).  Thus, bilateral focal lesions in the dorsal subparaventricular zone disrupts the circadian variation of body temperature, whereas bilateral lesions of the PVN, itself, is without effect (521).  Since the subparaventricular zone has prominent projections to the preoptic area (522), it is presumed that the suprachiasmatic nucleus relays information to thermosensitive neurons in the preoptic region by a multisynaptic pathway involving the subparaventricular zone (521).  However, the precise targets in the preoptic region from subparaventricular zone neurons have not yet been identified.

 

The set point for temperature regulation is also sensitive to circulating levels of sex steroids, with core body temperature falling just prior to the midcycle surge in women and rising during the luteal phase (523,524).  Estrogen, itself, would appear to be responsible for the fall in temperature in the late follicular phase by increasing the firing rate of preoptic warm sensitive neurons (525), whereas progesterone increases temperature in the luteal phase by decreasing the firing rate of preoptic warm sensitive neurons and, perhaps, increasing the firing rate of cold sensitive neurons (526).  Both steroids readily pass the blood-brain barrier and thereby are presumed to act directly on thermosensitive neurons in the preoptic nucleus (527).  Not unexpectedly, therefore, the lack of estrogen in the postmenopausal period gives rise to altered thermoregulatory responses (hot flashes) that occurs in over 80% of this group, and can be readily reversed by the administration of estrogen.  It has been proposed that in the absence of estrogen, the sensitivity of warm sensitive neurons to even small increases in body temperature are increased (528). Along these lines, it is of interest that in one study, the frequency of hot flashes was greatest during the afternoon and evening when body temperature normally rises under the influence of the suprachiasmatic nucleus circadian pacemaker (529).  The therapeutic response of some women with postmenopausal hot flashes to clonidine (530), an α2-adrenergic agonist, would indicate that catecholamines also participate in the heat loss responses.  Recent evidence, however, points to neurokinin B derived from kisspeptin (KNDy) neurons as having an important role. Kisspeptin neurons project to thermosensitive preoptic neurons that express neurokinin 3 receptors (NK3R), and when a NK3R agonist is administered into the proeptic area or neurokinin administered peripherally to human subjects, heat dissipation responses are observed (531,532).  Conversely, administration of an oral NK3R antagonist to postmenopausal women reduces the frequency and severity of hot flashes (533, 534).

 

A number of other substances have also been shown to modulate thermoregulatory responses, with thyroid hormone being one of the best studied examples and eluded to above.  Thyroid hormone is essential to sustain the effect of sympathetic activation of brown adipose tissue to produce heat.  Norepinephrine increases type 2 iodothyronine deiodinase in brown adipose tissue, which by converting T4 to T3 increases UCP-1 in mitochondria to convert stored energy into heat.  Although best known for its effects on arousal, orexin-A also is involved in the regulation of body temperature through effects on brown adipose tissue thermogenesis by increasing sympathetic activity and may explain dysregulation of thermogenesis in individuals with narcolepsy (535).  A role for prostaglandins in the febrile response associated with infection is discussed below.

 

Under certain circumstances, there is an adaptive advantage to elevate body temperature beyond the normal physiologic range that is highly conserved among animal species.  Such is the situation during infection, when fever is a necessary response to facilitate recovery by improving the efficiency of immune cells and impairing replication of microorganisms (536,537).  This homeostatic response is achieved by altering the thermoregulatory set point in medial preoptic neurons, but through a different mechanism than described above.  Under these circumstances, it is proposed that circulating endotoxin and proinflammatory cytokines interact with specific receptors on vascular endothelial cells and/or subendothelial microglia in the OVLT, resulting in activation of cyclooxygenase and production of PGE2 (538.539).  PGE2 released into the surrounding tissue binds to neighboring warm sensitive neurons in the median preoptic nucleus that express EP3 prostaglandin receptors (540), which by reducing GABAergic inhibition of thermogenic neurons in the hypothalamic paraventricular and dorsomedial nuclei and/or brainstem rostral ventromedial medulla (rMR, comprised of serotonergic neurons in the raphe pallidus and magnus, sympathetic premotor neurons), influence sympathetic preganglionic neurons in the spinal intermediolateral cell column that contribute to the generation of fever, cutaneous vasomotion, tachycardia and shivering (515, 541, 542).  The proposed mechanism is schematized in Fig 39.

 

Figure 39. Simplified diagram of major loci and pathways of the temperature control center. DH=dorsal horn, DMV=dorsomedial hypothalamus, IML=intermediolateral cell column, LPBN=lateral parabrachial nucleus, POA=preoptic area, PVN=paraventricular nucleus, rRPa=rostral raphe pallidus, VH=ventral horn. (Adapted from Roth J and Blasties CM, Mechanisms of fever production and lysis: Lessons from experimental LPS fever, Comp Physio 2014;4:1563-1604, 2014 and Lechan RM, Neuroendocrinology, in Williams Textbook of Endocrinology, 14th Edition, Hypothalamus and Pituitary, Ch 7, S. Melmed, R Auch us, A gold fine, RJ Koenig, C Fose, Eds, Elsevier, 2019.)

Blasties et al (543) suggest an alternative mechanism for fever induction in which PGE2 release into the preoptic region is mediated by norepinephrine, arising in noradrenergic (A2 cell group) neurons in the ventrolateral medulla.  This is based on the observation that in guinea pigs, the intra-preoptic microdialysis of α2-receptor antagonists potentiate the febrile response to LPS (544).   The mechanism proposed involves activation of hepatic branches of the vagus nerve by mediators (possibly PGE2) produced by liver Kupffer cells following the systemic administration of LPS.  Vagal afferent signals are then carried to the nucleus tractus solitarius in the brainstem, and after projecting to noradrenergic neurons in the ventrolateral medulla (A2), ascend in the ventral noradrenergic pathway and medial forebrain bundle to terminate in the preoptic region.  Ek et al (544) have also demonstrated in the rat that the intravenous administration of interleukin-1 is capable of activating vagal sensory neurons in the nodose ganglion and can be attenuated by inhibitors of prostaglandin synthesis.

 

In addition to inducing fever, endotoxin simultaneously activates an endogenous, counterregulatory, antipyretic response, to prevent body temperature from rising too severely.  This is largely achieved by stimulating the hypothalamic-pituitary-adrenal axis (see above) that exerts a dampening effect on the cytokine response, but more specifically by the direct antipyretic actions of α-MSH within the CNS (545).  The latter situation occurs only in association with cytokine activation, as α-MSH has no effect on temperature regulation in the absence of fever (545,546).  Alpha-MSH arises from the neuronal population in the hypothalamic arcuate nucleus (288), and while it is unknown precisely where α-MSH exerts its actions, the preoptic region including the VMPO is heavily innervated by axon terminals containing -MSH, suggesting a direct effect on thermosensitive neurons (547).  Alpha-MSH is also contained in axons that heavily innervate autonomic regulatory neurons in the parvocellular PVN and the hypothalamic DMN, providing an alternative route for regulatory control over vasomotor responses and heat generation.

   

Circadian Rhythmicity

 

Circadian rhythms are genetically determined, cyclic modifications of specific physiological functions and behaviors, generated through endogenous mechanisms in nearly all living organisms (548,549).  The basic organization of the circadian timing system includes an endogenous rhythm generator or pacemaker (also called endogenous clock or zeitgeber), a light-dark receptive system to entrain the endogenous clock to the time of day mediated by retinal photoreceptors (mainly cones) and visual pathways (retinohypothalamic pathway), and an efferent neural system coupling the pacemaker activity with effector systems in the brain that give rise to specific physiological functions and behaviors (548,549).  The master clock in mammals is the hypothalamic suprachiasmatic nucleus (SCN), a small, paired nucleus embedded in the dorsal surface of the optic chiasm.  Contained within this nucleus are multiple, small neurons that produce autonomous, self-sustaining oscillations synchronously firing to generate a common rhythmic output, perhaps mediated by the local release of GABA (550,551).  If the SCN is lesioned bilaterally, “free-running circadian rhythmicity” is produced, characterized by disruption of the sleep-wake cycle and loss of predictable daily oscillations in feeding, drinking, melatonin secretion and the secretion of some anterior pituitary hormones (552,553). Normal rhythmicity can be restored if the SCN is transplanted back into the lesioned animals (554).

 

Molecular mechanisms for the endogenous pacemaker activity of SCN neurons have been attributed to clock genes that include period (per), Clock, Cryptochrome (Cry), and Bmal (548,549).  Circadian oscillations in a number of other gene products mediated by microRNA (miR) have also been described in other tissues such as the liver (miR-122), retina (miR-26) and brain circuitries involved in locomotion (miR279) without any requirement for miRNA rhythmicity.  Presumably this mechanism involves translational regulation of clock protein mRNA by miR accumulation, and/or functional heterogeneity of miRNA species in a single, constitutively expressed miR population (555).  In addition, beyond transcriptional-translational feed-back loops occurring intracellularly, increasing evidence suggests that timekeeping mechanisms can be also controlled through non-transcriptional oscillators (NTO).  Examples of NTO in mammals are the redox based rhythms in post-translational modification of the antioxidant peroxiredoxin (PRX) proteins found in mouse liver and human red blood cells, suggesting that NTO can be coupled to classical transcriptional-translational feedback loops to regulate circadian rhythmicity (556).

 

Two different subdivisions of the SCN have been described, a ventrolateral and dorsomedial subdivision (548).  The ventrolateral subdivision or “core”, receives the major input to the SCN, including a massive projection of pituitary adenyl cyclase-activating peptide (PACAP)- and nitric oxide (NO)-containing axons from the retinohypothalamic pathway, GABA- and NPY-containing axonal projections from the intergeniculate leaflet of the thalamus, and serotonin neurons from the midbrain raphe (548,549).  These inputs have an important role in modulating the endogenous rhythms of the individual SCN pacemaker cells during the day / night alternance and as a result of changes in locomotor activity (557).  The dorsomedial subdivision or “shell”, primarily serves as the field for afferent information coming from the limbic system (hippocampus, bed nucleus of the stria terminalis, septum) and the hypothalamus, itself.  It is likely that through these inputs, cognitive and emotional information may exert phase-shifting effects on SCN pacemaker activity (528).  Both subdivisions are composed of a heterogeneous population of immunocytochemically distinct neurons.  The ventrolateral SCN contains neurons that express vasoactive intestinal polypeptide (VIP), gastric-releasing peptide and GABA (558).  The VIP-containing population seems to play a role in coordinating the different SCN neuronal groups involved in entraining different cyclic activities, thus ensuring intra-SCN synchrony (548).  Dorsomedial neurons express arginine vasopressin (AVP), angiotensin II, somatostatin, GABA (548) and prokineticin 2 (PK2) (559).  PK2 neurons have a distribution similar to AVP cells, and would be active during the light phase to favor locomotor patterns and thermoregulation, whereas they would remain silent at night (560).  However, while ventrolateral subdivision neurons receive light information, most of these neurons do not produce rhythmic patterns (561).  In contrast, the dorsomedial subdivision does contain rhythmic neurons, particularly apparent for AVP-producing neurons in which the peptide peaks during the day and is lowest at night (562).  This rhythmic pattern is partly secondary to the presence of binding sites for clock genes in the AVP promoter region (563), but also dependent upon synaptic transmission from other SCN neurons (564), perhaps those in the ventrolateral subdivision through intra-SCN connections (565) where VIP-containing cells are the best candidates (559).

 

The SCN has massive projections to three major regions of the diencephalon.  The most important is the hypothalamic subparaventricular zone (SPVZ).  Projections from the dorsal SPVZ reach the medial preoptic hypothalamus and are involved in the regulation of body temperature set-point and food-dependent energy intake (558).  In contrast, projections from the ventral SPVZ heavily innervate the hypothalamic dorsomedial nucleus and, to lesser extent, the midline thalamus, midbrain reticular formation and basal forebrain.  These outputs entrain photic stimuli with changes in food intake, rest/locomotion behavior, sleep-wake phases and pituitary hormone secretions. The second major projection is a GABAergic fiber tract to the hypothalamic PVN, and is primarily related to the secretion of melatonin from the pineal gland (558,566,567) by way of a multisynaptic pathway involving dorsal parvocellular neurons in the PVN, preganglionic cholinergic neurons in the intermediolateral cell column of the spinal cord, and postganglionic noradrenergic neurons in the superior cervical ganglion (567,568).  Melatonin is of importance as a humoral signal that feeds back on the SCN through melatonin receptors expressed in this nucleus, to facilitate sleep onset by communicating information concerning initiation and length of the dark phase (450,452). In addition, it regulates immune function, is of particular importance for reproductive activity in animals with seasonal breeding patterns (569,570), and participates in accommodating pituitary function with the shift to torpor in hibernators (571,572).  In homeothermic mammals, melatonin has been implicated in the inhibition of pituitary TSH biosynthesis at night (559).  A direct projection to the PVN may also mediate the day / night shift of anterior pituitary hormone secretion.  In rats, the SCN inhibits hypophysiotrophic CRH release through excitatory AVP outputs impinging onto GABAergic PVN interneurons.  In contrast, through direct SCN AVP outputs to tuberoinfundibular CRH cells (and possibly glutammateric inputs), release of CRH is triggered just before awaking, establishing circadian oscillation in the ACTH-glucocorticoid axis.  At the same time, autonomic PVN neurons are activated to stimulate the downstream, spinal, sympathetic outflow to the adrenal gland, resulting in increased ACTH sensitivity of the zona fasciculata (573,574).  However, circadian oscillations in CRH, ACTH and glucocorticoid release can also be regulated by the endogenous clock genes, per1, per2 and Bmal1, that exhibit antiphasic patterns in the PVN and adrenal cortex with respect to the pituitary corticotrophes.  The expression of these three, clock genes, in fact, can be differentially influenced by restricted feeding, acting as a metabolic effector independent of SCN outputs (575).  Similar to the sympathetic projections to the adrenal, the thyroid gland is also believed to receive an indirect SCN input via PVN neurons, activating sympathetic spinal outflow that would contribute to circadian regulation of thyroid hormone secretion (559).  A third major projection is directed to the medial and lateral tuberal hypothalamus, primarily the ventromedial nucleus, arcuate nucleus and lateral hypothalamic area.  These fibers are believed to influence the regulation of the neuroendocrine tuberoinfundibular and neurohypophysial secretions.  Figure 40 schematically depicts an integrated view of the mammalian circadian timing system and the main physiological functions and behaviors it controls.

Figure 40. Schematic drawing showing an integrated view of the mammalian circadian timing system and the main neuroendocrine responses, physiological functions and behaviors under its control. AII = angiotensin II; APv = anterior periventricular nucleus of the hypothalamus; AVP = arginine vasopressin; BNST = bed nucleus of the stria terminalis; CAL = calretinin; ENK = enkephalin; GABA = aminobutyric acid; GRP = gastrin-releasing peptide; GLU = glutamic acid; 5-HT = 5-hydroxytryptamine or serotonin; HYP = hypothalamus, IGL = intergeniculate leaflet; NO = nitric oxide; NPY = neuropeptide Y; PACAP = pituitary adenylyl cyclase-activating peptide; PK2= prokineticin 2; PTA = pretectal area; PVN = hypothalamic paraventricular nucleus; rht = retinohypothalamic tract; SCN = suprachiasmatic nucleus; SP = substance P; SPVZ = hypothalamic subparaventricular zone; SRIF = somatostatin; VIP = vasoactive intestinal polypeptide.

Sleep-Wake Cycle

 

Sleep is a natural state of altered consciousness, easily reversible, self-regulating and characterized by a stereotypic posture, decrease in voluntary motor activity and increase in arousal threshold.  In mammals and man, sleep periods are cyclically coupled to periods of wakefulness, giving rise to a circadian sleep-wake cycle.  Electrophysiologically, sleep is characterized by a progressively slower, higher voltage, and more synchronized electrical activity of the cortex (alpha waves – stage 1, spindle and k-complexes – stage 2, delta waves – stages 3 and 4) as opposed to wakefulness where fast, low voltage, and desynchronized electrical activity prevails (beta waves).  Only relatively brief times are spent in sleep-wake transitions.  Episodes of partial arousal without wakefulness occur during sleep, and are characterized by desynchronized electrical cortical activity resembling the EEG pattern of wakefulness and the initial sleep phase (theta waves).  This arousal is coupled to rapid eye movements (REM) and loss of muscle tone (except for respiratory and inner ear muscles).  In contrast, non-REM or NREM sleep is devoid of involuntary eye movements and muscle tone resumes, leading to deep sleep (576).

 

The basic neural organization of the sleep-wake cycle relies on two adjacent areas of the neuraxis, the diencephalon-basal forebrain and the mid-rostral brainstem, collectively expressing two, stable, firing states to produce either rest or arousal, with a tendency to avoid intermediate conditions (flip-flop switch).  In this manner, inappropriate behavioral fluctuations that might endanger survival are avoided, favoring discrete and rapid changes between sleep and waking profiles (or between REM and NREM sleep) above a background of slow and continuous variations in circadian and homeostatic inputs (either photic or non-photic autonomic, endocrine-metabolic and immune stimuli) (576,577).

 

The neural structures participating in this flip-flop switch establish reciprocal, feedback circuitries and can be classified as sleep- and wakefulness-promoting centers, the latter including specific cell groups that trigger and shut off the REM arousal state during sleep.  The primary sleep center is localized in the preoptic hypothalamus, and involves GABA- and galanin-containing neurons in the ventrolateral preoptic nucleus or VLPO (578).  A secondary sleep center is located in the midline thalamus (visceral or limbic thalamus), primarily in the dorsomedial (579) and reticular (580) thalamic nuclei. 

 

Wakefulness centers are numerous and in large part belong to the ascending reticular activating system of Moruzzi and Magoun (581).  This system can be subdivided in two different components, including monoaminergic and cholinergic cell groups in the pontine and mesencephalic (or limbic midbrain area) reticular formation.  The monoaminergic neurons comprise the noradrenergic locus coeruleus (LC), the serotoninergic median and dorsal raphe (DR) and parabrachial nucleus (PBN) (including some neurons that co-contain glutamage), and the dopaminergic ventral periaqueductal grey (vPAG) in the tegmentum of the mesencephalon. The cholinergic neurons are located in the pontine tegmentum, specifically in the laterodorsal (LDT) and peduncolopontine tegmental (PPT) nuclei, respectively.  In addition to pontine and mesencephalic reticular nuclei, wakefulness centers also involve histaminergic neurons in the posterior hypothalamus (tuberomammillary nucleus or TMN), peptidergic cell groups (orexin/hypocretin and melanin-concentrating hormone or MCH) in lateral hypothalamic area (LHA)/perifornical area (PF), and cholinergic and GABAergic magnocellular neurons in the basal forebrain (nucleus basalis of Maynert in the substantia innominata, magnocellular preoptic nucleus, medial septal nucleus, nucleus of the diagonal band of Broca) (572).

 

Axons from the primary sleep center (VLPO) travel caudally towards the wakefulness centers of the posterior hypothalamus (TMN) and ponto-mesencephalic reticular formation (LC, DR) via the medial forebrain bundle (MFB) (582).  Their cells of origin can be traced primarily to the VLPO core (VLPOc), but also extend to the periphery of this nuclear group (VLPOex) that innervate the LC, DR and mesopontine tegmentum (LDT and PPT).  Synaptic contacts in the LC and DR are primarily established by galanin-containing and to a lesser extent, GABAergic VPLO inputs (583-586).  VPLO synaptic contacts in the LDT and PPT are made with interneurons, likely disinhibiting inhibitory influences originating in TMN, LC and DR.  The signals are then directed to the principal cholinergic cells of the mesopontine tegmentum (557,558).

 

Fibers from the secondary sleep center (thalamic DM and reticular nuclei) course either in the periventricular system or in the MFB (thalamic peduncle of the ansa peduncolaris) to reach the periventricular and lateral preoptic hypothalamus (582) and mesopontine tegmental nuclei (588), respectively. Some of these axons contain glutamate and may enter the corona radiata to innervate the prefrontal cortex, that reproject back to the lateral hypothalamus (588).

 

Axons from the wakefulness cell groups (ponto-mesencephalic, hypothalamic and telencephalic) enter the MFB and travel rostrally through 2 pathways.  The first is through a dorsal route, mainly provided by cholinergic mesopontine tegmental nuclei (LDT and PPT) and, to a lesser extent, by cholinergic and GABAergic basal forebrain neurons.  Axons innervate primarily the intralaminar and reticular thalamic nuclei, that diffusely reproject to the cortex.  The second is through a ventral route arising from aminergic/glutamatergic pontine and mesencephalic nuclei (LC, PC, DR-PBN, vPAG), aminergic (histamine) and peptidergic (orexin/hypocretin and MCH) cell groups in the posterior and lateral hypothalamus (TMN and LHA/PF, respectively), as well as by the majority of cholinergic and GABAergic basal forebrain neurons.  These fibers cross the lateral hypothalamus and basal forebrain to reach all cortical areas (576,589).  However, only the PBN and paracoeruleus (PC) glutamateric projections diffusely innervate the magnocellular nuclei in the basal forebrain (590).  Finally, peptidergic (orexin/hypocretin and MCH) neurons in the LHA/PF also enter the MFB to heavily innervate aminergic and cholinergic cell groups in the posterior hypothalamus (TMN), pons-mesencephalon (LC, DR) and pontine tegmentum (LDT, PPT), respectively (564).

 

Initiation of sleep and maintenance of deep sleep are driven by the VLPO, via inhibition of monoaminergic centers in the posterior hypothalamus (TMN) and ponto-mesencephalic reticular formation (primarily LC and DR).  Rodents with excitotoxic lesions of the VLPO show a reduction of NREM sleep that closely correlates with the loss of Fos-immunoreactive neurons in the VLPOc (586,587).  Indeed, pontine and mesencephalic reticular nuclei (including the vPAG) and the cholinergic basal forebrain cells are responsible for electrical brain resynchronization, arousal and the waking state, through their excitatory dorsal- and ventral-projecting axonal pathways to the cortex (591,592).  Specifically, glutamatergic projections from the PBN and PC, mediated by basal forebrain centers, play a key role in maintaining wakefulnees and REM sleep depending on activation or inhibition of the ascending monoaminergic arousal system (590).  Such a possibility is consistent with clinical evidence that in humans, long lasting coma and a persistent vegetative state correlate with hemorrhage in the PBN region (593).  In contrast, reactivation of the PBN-PC complex by lesions of the inhibitory, mesopontine tegmentum, may trigger dream-like states with intense visual experiences in awake subjects at inappropriate times (so called peduncolar hallucinosis), mimicking cortical REM-like behavior in a state of vigilance (594).

      

Orexin/hypocretin cells in the LHA/PF contribute to the waking state by stimulating the posterior hypothalamic (TMN) and ponto-mesencephalic (LC, DR) aminergic waking centers, thus reinforcing cortical arousal.  Excitatory orexin 1 and 2 receptors have been found in all brainstem and basal forebrain wakefulness centers (595,596).  Injection of orexin/hypocretin into these areas increases neuronal firing (597,598), while its administration into the preoptic hypothalamus presynaptically inhibits VLPO excitability (599).  In addition, gene knockout for orexin/hypocretin, mutations in the orexin 2 receptor gene or absence of CSF orexin are associated with narcolepsy in mammals and man (600-602).

 

The VLPO also stimulates cholinergic neurons in the LDT and PPT to induce REM bursts, thus favoring arousal without wakefulness.  In particular, outputs from LDT and PPT (and possibly from part of the basal forebrain cells) are excitatory to thalamic neurons projecting to the cortex.  When the VLPO activates the reticular pontotegmental nuclei, the transthalamic sensory transmission may easily diffuse to the cortical mantle, leading to enhancement of cortical arousal during conditions of synchronized brain activity (NREM to REM shift) (576,591).  Conversely, loss of Fos-immunoreactive neurons in the VLPOex correlates with a reduction in REM sleep episodes (594,595).

 

In contrast, cholinergic neurons in the basal forebrain (medial septum and diagonal band of Broca) are implicated in the generation of specific electrical activity during REM episodes (theta waves) in response to stimulation by brainstem aminergic inputs (603) such as from the PBN-PC complex (590).   Also, MCH neurons in the LHA/PF contribute to the REM sleep by inhibiting the ponto-mesencephalic monoaminergic inputs to the cortex.  This inhibition amplifies arousal without wakefulness (REM phase) mediated by mesopontine tegmental centers (LDT and PPT) (576).  A reciprocal, negative feedback circuitry is established between pontine aminergic cell groups (LC area and tegmental area), leading to episodic switch between NREM and REM phases (605).  Finally, thalamic DM and reticular neurons likely come into play to coordinate either REM or NREM sleep with other behavioral and endocrine regulations (605), as well as to increase arousal threshold by reinforcing the inhibitory action of the VLPO on rostral mesencephalic waking centers (580).  Consistently, excitotoxic lesions of the thalamic DM induce persistent insomnia in cats (606) and its degeneration in humans gives rise to fatal familial insomnia, a disorder characterized by loss of rhythmicity in sleep-wake cycle, body temperature, blood pressure and anterior pituitary secretions (607).  Figure 41 shows a simplified and integrated view of the neural circuitry controlling the mammalian sleep-wake cycle.

Figure 41. Schematic drawing showing a simplified and integrated view of the neural circuitry controlling the mammalian sleep-wake cycle. Different colors highlight the pathways for wakefulness (red and pink), NREM sleep (blue) and REM sleep (green). Continuous lines indicate primary circuitries for initiation of the sleep and waking state, dotted and hatched lines permissive circuitries for different sleep phases (NREM vs REM) and behavioral arousal, mixed colors different patterns of activity of the same center in relation to either sleep phases or wakefulness. Ach = acetylcholine; DA = dopamine; NDBB = nucleus of the diagonal band of Broca; DM = thalamic dorsomedial nucleus; DR = dorsal raphe; GABA =  aminobutyric acid; GAL = galanin; Glut = glutamic acid; His = histamine; 5HT = 5-hydroxytryptamine or serotonin; LC = locus coeruleus; LDT = laterodorsal tegmental nucleus; MCH = melanocortin-concentrating hormone; MPO = magnocellular preoptic nucleus; MSN = medial septal nucleus; NA = noradrenaline; NBM = nucleus basalis of Maynert; ORX = orexin/hypocretin; PBN = parabrachial nucleus; PPT = peduncolopontine tegmental nucleus; Reticular = thalamic reticular nucleus; vPGA = ventral periacqueductal grey; VPLO = hypothalamic ventrolateral preoptic nucleus, VPLOc = VPLO core; VPLOex = VPLO extended; TMN = hypothalamic tuberomammillary nucleus; + = stimulation; - = inhibition

Major regulators of the neural machinery for the sleep-wake cycle are the circadian timing system (CTS), feedback regulation exerted by locomotor activity and sleep-wake cycle itself onto the CTS, and homeostatic mechanisms endogenous to the sleep and wakefulness neural circuitries (sleep homeostat) (576) including the activity of the glymphatic system (212).  The CTS is primarily governed by the hypothalamic suprachiasmatic nucleus (SCN) and its widespread CNS projections, including those that trigger melatonin secretion from the pineal gland (see section F. Circadian Rhythmicity).  In rats, sleep-wake rhythmicity is adaptively coordinated with feeding behavior.  In fact, light sensitive, SCN projections to the ventral subparaventricular zone (608) innervate food-entrainable neurons in the hypothalamic dorsomedial nucleus (608,609) that sends stimulatory glutamate-containing inputs to LHA orexin neurons and inhibitory GABAergic inputs to VLPO neurons (610).  In this manner, photic-dependent stimuli can be integrated with nonphotic stimuli to establish a rest/sleep-locomotion/wakefulness cycle that is ideal for nutritional success (611).  In turn, locomotor activity may feed back onto the SCN by activating NPY-containing outputs in the intergeniculate leaflet of the thalamus and serotoninergic projections in the median raphe nucleus to entrain the sleep-wake cycle with levels of exploratory behavior (612).  In humans, disruption of this locomotor-circadian rhythm circuitry has been implicated as a potential cause for development of cognitive decline and neurodegeneration.  In support, recent evidence in Drosophila shows that stable, long-term memory depends on circadian cycles in rest and activity, leading to impairment in memory consolidation when sleep disruption occurs.  In fact, increased locomotion and waking state results in loss of synaptic remodeling at critical brain sites (613) such as the hypocretin/orexin system in the LHA where the number of synapses oscillates with light and dark phases (614).  In addition, stress has been associated with increased cFos expression in a number of limbic areas (infralimbic cortex, central nucleus of the amygdala, bed nucleus of the stria terminalis) projecting to both activating (VLPO) and inhibiting (LC, TMN) sleep centers.  These inputs may be important for maintaining a waking state during behavioral arousal, such as an emergency occurring during normal sleep. Their activation by anxiety may contribute to stress-induced insomnia (615).  Hyperactivation of corticolimbic sites has also been shown in human subjects with insomnia and might contribute to the excessive high frequency EEG activity seen during NREM sleep, including their sensation of being awake even when the EEG appears to be in NREM, a condition known as “sleep state misperception” (616).

 

Finally, some cellular and molecular mechanisms may serve as an internal homeostat to inhibit aminergic waking centers and activate the VLPO (613,617).  In particular, in conditions of sleep deprivation, rat astrocytes have been found to release adenosine in the perineuronal space of LC and orexigenic LHA/PF to shut off monoaminergic excitatory systems via A1 inhibitory adenosine receptors. This inhibition of noradrenergic arousal mechanisms would also favor activation of the hypothalamic glymphatic flow by increasing the interstial space, and relieving the restraining effect of catecholaminergic inputs to the choroid plexus, resulting in increased CSF production.  Thus, neurotoxic waste products could be more removed, and the brain neurons cleared as during a regular sleep (212). Extraneuronal adenosine may also reduce presynaptic inhibition of VLPO neurons via A2 excitatory adenosine receptors, thereby triggering compensatory sleep (613).

 

This and a number of circulating endocrine, metabolic and immune inputs controlled by the CTS may influence circadian clock genes (576,618) that in turn, influence the sleep-wake cycle.  In particular, knockout mice for Clock, Cry1/Cry2, and Bmal1 in the SCN and related neural pathways show either an increase (Clock) or decrease (Cry and Bmal1) in NREM sleep (619-621).  In rodents, Clock and Bmal1 deactivation also reduces orexin outputs in the LHA, giving rise to a hyperphagic, obese and dysmetabolic phenotype.  Other gene candidates involved in this regulation include REV-Erba1, whose impairment leads to altered locomotor activity in either constant light or darkness, and PGC-1α / PGC-1β, that control metabolic rate in liver and muscle, body temperature and locomotor activity in the dark phase (622).  Collectively, clock gene activation / deactivation phases by the CTS during the sleep-wake cycle may play a key role in regulating synaptic plasticity and metabolic balance.

 

REFERENCES

 

  1. Christ JF. 1969 Derivation and boundaries of the hypothalamus, with atlas of hypothalamic grisea, in Haymaker W, Anderson E, Nauta WJH (eds): The Hypothalamus. Charles C. Thomas, pp 13-60.
  2. May MT. 1968 Galen: On the usefulness of the parts of the body.De Usu Partium. Cornell University Press, pp 424- 461.
  3. Toni R.2000 Ancient views on the hypothalamic-pituitary-thyroid axis: an historical and epistemological perspective.  Pituitary 3: 83-95.
  4. Meshberger FL.1990 An interpretation of the Michelangelo’s Creation of Adam based on neuroanatomy.  JAMA 10: 1837-1841.
  5. His W.1893  Vorschlage zur Einteilung des Gehirns.  Arch Anat Physiol, Anat Alb 172-179.
  6. Toni R, Malaguti A, Benfenati F, Martini L. The human hypothalamus: a morphofunctional perspective. J Endocrinol Invest 27 (supp to n.6), 73-94, 2004.
  7. Ciocca DR, Puy LA, Stati AO. 1984 Constitution and behavior of follicular structures in the human anterior pituitary gland. Am J Pathol 115:165-174.

8.Horvath E, Kovacs K.  2002 Folliculo stellate cells of the human pituitary: a type of adult stem cells?  Ultrastruct Pathol 26: 219-228.

  1. Fitzgerald KT. 1979 The structure and function of the pars tuberalis of the vertebrate adenohypophysis. Gen Comp Endocrinol 37: 383-399.
  2. Cohen LE, Radovick S. 2002 Molecular basis of combined pituitary hormone deficiencies. Endocr Rev 23: 431-442.
  3. Zhu X, Gleiberman AS, Rosenfeld MG.2007 Molecular physiology of pituitary development: signaling and transcriptional networks.  Physiol Rev 87:  933-963.
  4. Zhu X, Wang J, Ju B-G, Rosenfeld MG. 2007 Signalling and epigenetic regulation of pituitary development. Curr Op Cell Biol 19: 605-611.
  5. Alatzoglou KS, Kelberman D, Dattani MT.2009 The role of SOX proteins in normal pituitary development.J. Endocrinol 200, 245–258.
  6. Nakane PK. 1970 Classifications of anterior pituitary cell types with immunoenzyme histochemistry. J Histochem Cytochem 18: 9-20.
  7. Snyder G, Hymer WC, Snyder J. 1977 Functional heterogeneity in somatotrophs isolated from the rat anterior pituitary. Endocrinology101: 788-799.
  8. Nikitovitch-Winer MB, Atkin J, Maley BE. 1987 Colocalization of prolactin and growth hormone within specific adenohypophyseal cells in male, female and lactating female rats. Endocrinology121: 625-630.
  9. Frawley LS, Clark CL, Schoderbek WE, Hoeffler JP, Boockfor FR. 1986 A novel bioassay for lactogenic activity: Demonstration that prolactin cells differ from one another in bio-and immuno-potentencies of secreted hormone. Endocrinology119: 2867-2869.
  10. Schwartz J, Canny B, Vale WW, Funder JW. 1989 Intrapituitary cell-cell communication regulates ACTH secretion, Neuroendocrinology 50: 716-722.
  11. Nakajima T, Yamaguchi H, Takahashi K. 1980 S100 protein in folliculostellate cells of the rat pituitary anterior lobe. Brain Res191: 523-531.
  12. Denef C. 1994 Paracrine mechanisms in the pituitary, in Imura H (ed): The Pituitary Gland. Raven Press, pp 351-378.
  13. Vankelecom H.2007 Non-hormonal cell types in the pituitary candidating for stem cell.  Sem in Cell Develop Biol 18: 559-570.
  14. Rizzoti K. 2010. Adult pituitary progenitors/stem cells: from in vitro characterization to in vivo function.Eu. J. Neurosci. 32: 2053–2062.
  15. Garcia-Lavandeira M, Quereda V, Flores I, Saez C, Diaz-Rodriguez E, Japon MA, Ryan AK, Blasco MA, Dieguez C, Malumbres M, Alvarez CV 2009 A GRFa2/Prop1/stem (GPS) cell niche in the pituitary. PloS One 4:e4815.
  16. Bergland RM, Page RB. 1978 Can the pituitary secrete directly to the brain? Endocrinology102:1325-1338.
  17. Leclercq T, Grisoli F. 1983 Arterial blood supply of the normal human pituitary gland. J Neurosurg 58: 678-681.
  18. Porter JC, Kamberi IA, Grazia YR. 1978 Pituitary blood flow and portal vesels, in Martini L, Ganong W (eds): Frontiers in Neuroendocrinology. Oxford University Press, pp 145-175.
  19. Gross PM, Joneja M, Pang JJ, Polischuk TM, Shavers SW, Wainman DS. 1993 Topography of short portal vessels in the rat pituitary gland: a scanning electron-microscopic and morphometric study of corrosion cast replicas. Cell Tiss Res 242: 79-88.
  20. Yuh WTC, Fisher DJ, Nguyen HD, Tali ET, Gao F, Simonson TM, Schlechte JA. 1994 Sequential MR enhancement pattern in normal pituitary gland and in pituitary adenoma. AJNR 15: 101-108.
  21. Wittkowski WH, Schulze-Bonhage AH, Bockers TM. 1992 The pars tuberalis of the hypophysis: A modulator of the pars distalis? Acta Endocrinologica126: 285-290.
  22. Iliff JJ, Lee H, Yu M, Feng T, Logan J, Nedergaard M, Benveniste H. 2013 Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. J Clin Invest. 123:1299-309.
  23. Altman J, Bayer SA. 1986 Development of the rat hypothalamus. Adv. Anat. Embriol. Cell Biol 85.
  24. Markakis E. A. 2002 Development of the neuroendocrine hypothalamus. Frontiers in Neuroendocrinoloty 23: 257-291. 
  25. Christ J. F. 1969 Derivation and boudaries of the hypothalamus, with atlas of hypothalamic grisea, in Haymaker W, Anderson E, Nauta WJH (eds): The Hypothalamus. Charles C. Thomas, pp 13-60.
  26. Wataya T, Ando S, Maguruma K, Ikeda H, Watanabe K, Eiraku M, Kawada M, Takahashi J, Hashimoto N, Sasai Y.2008 Minimization of exogenous signals in ES culture induces rostral hypothalamic differentiation.  Proc Natl Acad Sci 105: 11796-11801.
  27. Tessmar-Raible K, Raible F, Christodoutou F, Guy Keren, Rembold M, Hausen H, Arendt D.2007 Conserved sensory-neurosecretory cell types in annelid and fish forebrain:  Insights into hypothalamic evolution.  Cell 129: 1389-1400.
  28. Sauer F. C. 1935 Mitoses in the neural tube. J. Comp. Neurol. 62: 377-405.
  29. Ohyama K, Das R, Placzek M.2008 Temporal progression of hypothalamic patterning by a dual action of BMP Development 135, 3325-3331.
  30. Figdor MC, Stern CD. 1993 Segmetnal organization of the embryonic diencephalon. Nature 363: 630-634.
  31. Coggeshall RE. 1964 A study of diencephalic development in the albino rat. J. Comp. Neurol. 122: 241-270.
  32. Koutcherov Y, Mai JK, Ashwell KWS, Paxinos G.2002 Organization of human hypothalamus in fetal development.  J Comp Neurol 446: 301-324.
  33. Markakis EA, Swanson L.W. 1997 Spatiotemporal patterns of secretomotor neuron generation in the parvicellular neuroendocrine system.Brain Res. Rev. 24: 255-291.
  34. Schwanzel-Fukuda M, Pfaff DW. 1989 Origin of luteinizing hormone-releasing hormone neurons. Nature 338: 161-164.
  35. Toni R, Briganti F., Lechan RM, Vezzadini P. 1996 Immunocytochemical distribution of somatostatin in the hypothalamus of the human fetus. Abstracts 10th International Congress of Endocrinology, S. Francisco, p.292.
  36. Toni R, Musolino A, Ruggeri F, Scialpi C. 1996 Immunocytochemical distribution of thyrotropin-releasing hormone (TRH) prohormone in the diencephalon of the human fetus. It. J. Anat. Embriol. 101 (supp):2.
  37. Alvarez-Bolado G, Rosenfeld MG, Swanson LW. 1995 Models of forebrain regionalization based on spatiotemporal patterns of POU-III homeobox gene expression, birthdates and morphological features. J. Comp. Neurol. 355: 237-295.
  38. Nakai S, Kawano H, Yudate T, Nishi M, Kuno J, Nagata A, Jishage K, Hamada H, Fujii H, Kawamura K, Shiba K, Noda T. 1995 The POU domain transcription factor Brn-2 is required for the determination of thedevelopmen of the rat somatostatin-containing neuocrotical neuronal system. Genes Dev.  9: 3109-3121.
  39. Schonemann MD, Ryan A K, McEvilly RJ, O’Connell SM, Arias CA, Kalla KA, Li P, Sawchenko PE, Rosenfeld MG. 1995 Development and survival of the endocrine hypothalamus and posterior pituitary glnd requires the neuronal POU domain factro Brn-“. Genes Dev.  9: 3122-3135.
  40. Michaud J.L., Rosenquist T, May NR, Fan C-M. 1998 Development of neuroendocrine lineages require the bHLH-PAS transcription factor SIM1. Genes Dev. 12: 3264-3275.
  41. Norsted E, Gomuc B, Meister B. 2008 Protein components of the blood-brain barrier (BBB) in the mediobasal hypothalamus, J Chem Neuroanat. 36: 107-121.
  42. Michaud JL, DeRossi C, May NR, Holdener BC, Fan C-M. 2000 ARNT2 acts as the dimerization partner of SIM1 for the development of the hypothalamus. Mech Dev. 90: 253-261.
  43. lle Duplan SM, Bouche F, Alexandrov L, Michaud JL. 2009 Impact of Sim1 gene dosage on the development of the paraventricular and supraoptic nuclei of the hypothalamus.Eu. J. Neurosci. 2239–2249, 2009.
  44. Tobet ST.2002 Genes controlling hypothalamic development and sexual differentiation.  Eu J Neurosci 16: 373-376.
  45. Kurrasch DM, Cheung CC, Lee FY, Tran PV, Hata K, Ingraham HA.2007 The neonatal ventromedial hypothalamus transcriptome reveals novel markers with spatially distinct pattering.  J Neurosci 27: 13624-13634.
  46. Ivanova E, Kelsey G. 2011 Imprinted genes and hypothalamic function.J Mol Endocrinol 47; R67-R74.
  47. Swanson L.W. 1987 The hypothalamus, in Bjorklund A. Hokfelt T, Swanson LW (eds), Handbook of Chemical Neuroanatomy, vol 5, Integrated Systems of the CNS, part 1, Elsevier Science Publisher, pp 1-124.
  48. Thomson RH, Swanson LW. 2003 Structral characterization of a hypothalamic visceromotor pattern generator network.Brain Res Rev 41: 153-202. 
  49. Weindl A. 1973 Neuroendocrine aspects of circumventricular organs, in Ganong W, Martini L (eds): Frontiers in Neuroendocrinology, Oxford University Press, pp 3-32.
  50. Oldfield BJ, McKinley MJ. 1995 Circumventricular organs in Paxinos G (ed) The Rat Nervous System, Academic Press, pp 391-403.
  51. Johnson AK, Gross PM. 1993 Sensory circumventricular organs and brain homeostatic pathways. FASEB J 7: 678-686.
  52. Monroe BG. 1967 A comparative study of the ultrastructure of the median eminence, infundibular stem and neural lobe of the hypophysis of the rat. Zeitschrift fur Zellforschung 76: 405-432.
  53. Knigge KM, Scott DE. 1970 Structure and function of the median eminence. Am J Anat 129: 223-244.
  54. Ugrumov MV. 1992 Development of the median eminence during ontogenesis (morpho-functional aspects). in Ermisch A, Landgraf R, Ruhle H-J (eds): Progress in Brain Research, vol 91, pp 349-356.
  55. Petrov T, Howarth AG, Krukoff TL, Stevenson BR. 1994 Distribution of the tight junction-associated protein ZO-1 in circumventricular organs of the CNS. Mol Brain Res 21: 235-246.
  56. Krisch B, Leonhardt H. 1978 The functional and structural border of the neurohemal region of the median eminence. Cell Tissue Res 192: 327-339.
  57. Rodriguez EM, Blazquez JL, Pastor FE, Pelaez B, Peruzzo B, Amat P. 2005. Hypothalamic tanycytes: a key component of brain-endocrine interaction. Int Rev cytol 247: 89-164.
  58. Lechan RM, Fekete C. 2007 Infundibular tanycytes as modulators of neuroendocrine function: hypothetical role in the regulation of the thyroid and gonadal axis. Acta Biomed 78: 84-98.
  59. Wittmann G, Farkas E, Szilvasy-Szabo A, Gereben B, Fekete C, Lechan RM. 2016. Variable proopiomelanocortin expression in tanycytes of the adult rat hypothalamus and pituitary stalk. J Comp Neurol, in press.
  60. Scott DE, Paull WK. 1979 The tanycyte of the rat median eminence. Cell Tissue Res 200: 329-334.
  61. King J, Rubin BS. 1994 Dynamic changes in LHRH neurovascular terminals with various endocrine conditions in adults. Hormones and Behavior 28:349-356.
  62. Yoshimura T, Yasuo S, Watanabe M, Iigo M, Yamamura T, Hirunagi K, Ebihara S. 2003 Light-induced hormone conversion of T4 to T3 regulates photoperiodic response iof gonads in birds. Nature 13: 178-181.
  63. Beagley GH, Hatton GI. 1992 Rapid morphological changes in supraoptic nucleus and posterior pituitary induced by a single hypertonic saline injection. Brain Res Bull 28: 613-618.
  64. Ondo JG, Mical RS, Porter JC. 1972 Passage of radioactive substances from CSF to hypophysial portal blood. Endocrinology 91: 1239-1246.
  65. Kendall JW, Jacobs JJ, Kramer RM. 1972 Studies on the transport of hormones from the cerebrospinal fluid to hypothalamus and pituitary. in Knigge KM, Scott DE, Weindl A (eds) Brain-Endocrine Interaction. Median Eminence: Structure and Function, Karger, Basel, pp. 342-349.
  66. Kozlowski GP, Coates PW. 1985 Ependymoneural specializations between LHRH fibers and cells of the cerebroventricular system. Cell Tiss Res 242: 301-311.
  67. Tu HM, Kim S-W, Salvatore D, Bartha T, Legradi G, Larsen PR, Lechan RM. 1997 Regional distribution of type 2 thyroxine deiodinase messenger ribonucleic acid in rat hypothalamus and pituitary and its regulation by thyroid hormone. Endocrinology 138: 3359-3368.
  68. Fekete C, Freitas BC, Zeold A, Wittmann G, Kadar A, Liposits Z, Christoffolete MA, Singru P, Lechan RM, Bianco AC, Gereben B. 2007 Expression patterns of WSB-1 and USP-33 underlie cell-specific posttranslational control of type 2 deiodinase in the rat brain. Endocrinology 148: 4865-4874.
  69. Kallo I, Mohacsik P, Vida B, Zeold A, Bardoczi Z, Zavacki AM, Farkas E, Kadar A, Hrabovszky E, Arrojo E, Drigo R, Dong L, Barna L, Palkovitz M, Borsay BA, Herczeg L, Lechan RM, Bianco AC, Liposits Z, Fekete C, Gereen B. 2012 A novel pathway regulates thyroid hormone availability in rat and human hypothalamic neurosecretory neurons. PLoS One 7: e37860.
  70. Sanchez, E., M. A. Vargas, P. S. Singru, I. Pascual, F. Romero, C. Fekete, J. L. Charli and R. M. Lechan (2009). "Tanycyte pyroglutamyl peptidase II contributes to regulation of the hypothalamic-pituitary-thyroid axis through glial-axonal associations in the median eminence." Endocrinology 150(5): 2283-2291.
  71. Freitas, B. C., B. Gereben, M. Castillo, I. Kallo, A. Zeold, P. Egri, Z. Liposits, A. M. Zavacki, R. M. Maciel, S. Jo, P. Singru, E. Sanchez, R. M. Lechan and A. C. Bianco (2010). "Paracrine signaling by glial cell-derived triiodothyronine activates neuronal gene expression in the rodent brain and human cells." J Clin Invest 120(6): 2206-2217;
  72. Ebling, F. J. P. and J. E. Lewis (2018). "Tanycytes and hypothalamic control of energy metabolism." Glia 66(6): 1176-1184
  73. Farkas, E., E. Varga, B. Kovacs, A. Szilvasy-Szabo, A. Cote-Velez, Z. Peterfi, M. Matziari, M. Toth, D. Zelena, Z. Mezricky, A. Kadar, D. Kovari, M. Watanabe, M. Kano, K. Mackie, B. Rozsa, Y. Ruska, B. Toth, Z. Mate, F. Erdelvi, G. Szabo, B. Gereben, R. M. Lechan, J. L. Charli, P. Joseph-Bravo and C. Fekete (2020). "A glial-neuronal circuit in the median eminence regulates thyrotropin-releasing hormone-release via the endocannabinoid system." iScience 23(3): 100921.
  74. Lee DA, Bedoni JL, Pak T, Wang H, Song J, Miranda-Anquio A, Takiar V, Charubhumi V, Balordi F, Takebayashi H, Aja S, Ford E, Fishell G, Blackshaw S. 2012. Tanycytes of the hypothalamic median eminence form a diet-responsive neurogenic niche. Nat Neurosci 15: 700-702.
  75. Sanders NM, Dunn-Meynell AA, Levin BE. 2004. Third ventricular alloxan reversible impairs glucose counterregulatory responses.Diabetes 53: 1230-1236.
  76. Wittmann G, Farkas E, Szilvasy-Szabo A, Gereben, Fekete C, Lechan RM, Variable proopiomelanocortin expression in tanycytes of the adult rat hypothalamus and pituitary stalk. J Comp Neurol, in press.
  77. Robins SC, Stewart I, McNay DE, Taylor V, Giachino C, Goetz M, Ninkovic J, Briancon N, Maratos-Flier E, Flier JS, Kokoeva MV, Placzek M. 2013. A-Tanycytes of the adult hypothalamic third ventricle include distinct populations of FGF-responsive neural progenitors. Nat Commun 4: 1-13.
  78. Clarke I, Jessop D, Millar R, Morris M, Bloom S, Lightman S, Cohen CW, Lew R, Smith I. 1993 Many peptides that are present in the external zone of the median eminence are not secreted into the hypophysial portal blood of sheep. Neuroendocrinology 57:765-775.
  79. Merchenthaler I, Gorcs T, Setalo G, Petrusz P, Flerko B. 1984 Gonadotropin-releasing hormone (GnRH) neurons and pathways in the rat brain. Cell Tissue Res 237: 15-29.
  80. Kuljis RO, Davis JP. 1989 Immunocytochemical and physiological evidence of synapse between dopamine- and luteinizing-hormone releasing hormone-containing neurons in the ewe median eminence. Endocrinology124: 1579-1581.
  81. Contijoch AM, Gonzalez C, Singh HN, Malamed S, Troncoso S, Advis J-P. 1992 Dopaminergic regulation of luteinizing hormone-releasing hormone release at the median eminence level: Immunocytochemical and physiological evidence in hens. Neuroendocrinology 55:290-300.
  82. Sahu A, Crowley WR, Tatemoto K, Balasubramaniam A, Kalra SP. 1987 Effects of neuropeptide Y, NPY analog (norleucine4-NOY), galanin and neuropeptide K on LH release in ovariectomized (OVX) and OVX estrogens, progesterone-treated rats. Peptides 8:921-926.
  83. Lopez FJ, Negro-Vilar A. 1990 Galanin stimulates luteinizing hormone-releasing hormone secretion from arcuate nucleus-median eminence fragments in vitro: Involvement of an alpha-adrenergic mechanism. Endocrinology 127:2431-2436.
  84. Gross PM. 1992 Circumventricular organ capillaries. Prog Brain Res 1:219-233.
  85. Sutton SW, Toyama TT, Otto S, Plotsky PM. 1988 Evidence that neuropeptide Y (NPY) released into the hypophysial-portal circulation participates in priming gonadotropes to the effects of gonadotropin releasing hormone (GnRH). Endocrinology 123:1208-1210.
  86. Sahu A, Crowley WR, Kalra SP. 1994 Hypothalamic neuropeptide-Y gene expression increases before the onset of the ovarian steroid-induced luteinizing hormone surge. Endocrinology 134:1018-1022.
  87. Mezey E, Kiss JZ. 1985 Vasoactive intestinal polypeptide containing neurons in the paraventricular nucleus may participate in regulating prolactin secretion. Proc Natl Acad Sci USA 82:245-247.
  88. Stillman MA, Recht LD, Rosario SL, Seif SM, Robinson AG, Zimmerman EA. 1977 The effects sof adrenalectomy and glucocorticoid replacement on vasopressin and vasopressin-neurophysin in the zona externa of the median eminence of the rat. Endocrinology 101: 42-49.
  89. King J, Letourneau RJ. 1994 Luteinizing hormone-releasing hormone terminals in the median eminence of rats undergo dramatic changes after gonadectomy, as revealed by electron microscopic image analysis. Endocrinology 1994; 134: 1340-1351.
  90. Villar MJ, Meister B, Hokfelt T. 1994 Reorganization of neural peptidergic systems in the median eminence after hypophysectomy. J Neurosci 14:5996-6012.
  91. Liao N, Bulant M, Nicolas P, Vaudry H, Pelletier G. 1988 Electron microscope immunocytochemical localization of thyrotropin-releasing hormone (TRH) prohormone in the rat hypothalamus. Neuropeptides 11:107-110.
  92. Bulant M, Roussel J-P, Astier H, Nicolas P, Vaudry H. 1990 Processing of thyrotropin-releasing hormone prohormone (pro-TRH) generates a biologically active pepide, prepro-TRH-(160-169), whch regulates TRH-induced thyrotropin secretion. Proc Natl Acad Sci USA 87:4439-4443.
  93. Niimi M, Takahara J, Sato M, Kawanishi K. 1990 Immunohistochemical identification of galanin and growth hormone-releasing factor-containing neurons projecting to the median eminence of the rat. Neuroendocrinology 51:572-575.
  94. Ottlecz A, Samson WK, McCann SM. 1986 Galanin: Evidence for a hypothalamic site of action to release growth hormone. Peptides 7:51-53
  95. Davis TME, Burrin JM, Bloom SR. 1987 Growth hormone (GH) release in response to GH-releasing hormone in man is 3-fold enhanced by galanin. J Clin Endocrinol Metab 65:1248-1252.
  96. Lechan RM, Wu P, Jackson IMD, Wolfe H, Cooperman S, Mandel G, Goodman RH. 1986 Thyrotropin-releasing hormone precursor: Characterization in rat brain. Science 231:159-161.
  97. Ceccatelli S, Eriksson M, Hokfelt T. 1989 Distribution and coexistence of corticotropin-releasing factor-, neurotensin-, enkephalin-, cholecystokinin-, galanin- and isoleucine-like peptides in the parvocellular part of the paraventricular nucleus. Neuroendocrinology 9:309-323.
  98. Hokfelt T, Fahrenkrug J, Ju G, Ceccatelli S, Tsuruo Y, Meister B, Mutt V, Rundgren M, Brodin E, Terenius L, Hulting A-L, Werner S, Bjorklund H, Vale W. 1987 Analysis of peptide histidine-isoleucine and vasoactive intestinal polypeptide-immunoreactive neurons in the central nervous system with special reference to their relation to CRF- and enkephalin-like immunoreactivities in the paraventricular hypothalamic nucleus. Neuroscience 23: 827-857.
  99. Ceccatelli S, Lundberg JM, Fahrenkrug J, Bredt DS, Snyder SH, Hokfelt T. 1992 Evidence for involvement of nitric oxide in the regulation of hypothalamic portal blood flow. Neuroscience 51:769-772.
  100. Ceccatelli S, Fahrenkrug J, Villar MJ, Hokfelt T. 1991 Vasoactive intestinal polypeptide/peptide histidine isoleucine immunoreactive neuron systems in the basal hypothalamus of the rat with special reference to the portal vasculature: An immunohistochemical and in situ hybridization study. Neuroscience 43:483-502.
  101. Said SI, Mutt V. 1970 Polypeptide with broad biological activity: Isolation from small intestine. Science 169:1217-1218.
  102. Palmer RMJ, Ferrige AG, Moncada S. 1987 Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 327:524-526.
  103. Weindl A., Joynt R., Rochester N. 1972 Ultrastructure of the ventricular walls. Ach. Neurol (Chicago) 26: 420-426.
  104. Mitro A, Palkovits M. 1981 Morphology of the rat brain ventricles, ependyma and periventricular structures. Bibliotheca Anatomica, 21, S. Karger.
  105. Del Brio MA, Riera P, Gracia JM, Alvariz-Uria M. 1990 Ultrastructural study of the cellular components of the organum vasculosum of the lamina terminalis in the rabbit (Oryctolagus cuniculus). J. Submicrosc. Cytol. Pathol. 22: 303-309.
  106. Nakay Y, Kudo J, Hashimoto A. 1980 Specific cell membrane differentiation in the tanycytes and glial cells of the organum vasculosum of the lamina terminalis in dog. J. Electron Microsc. 29: 144-150.
  107. King J.C. Anthony ELP. 1984 LHRH neurons and their projections in humans and other mammals: Species comparisons. Peptides (Supp 1), 195-207.
  108. Yamaguchi K, Morimoto A, Murakami N. 1993 Organum vasculosum laminae terminalis (OVLT) in rabbit and rat: topographic studies. J. Comp. Neurol. 330: 352-362.
  109. Weindl A, Sofroniew MV. 1981 Relation of neuropeptides to mammalian circumventricular organs, in Martin JB, Reichlin S, Bick KL (eds): Neurosecretion and Brain Peptides, Raven Press, pp. 303-320.
  110. Palkovits M, Mezey E, Ambach G, Kivoviks P. 1978 Neural and vascular connections between the organum vasculosum laminae terminalis and preoptic nuclei. Brain-Endocrine Interaction IIII. Neural Hormones and Reproduction, D.E. Scott, G.P. Kozlowski, A. Weindl, Eds, Karger, Basal, pp 63-65.
  111. Ciura S, Bourque CW. 2006 Transient receptor potential vanilloid 1 is required for intrinsic osmoreception in organum vasculosum lamina terminalis neurons and for normal thirst responses to systemic hyperosmolality. J Neuroscience 26: 9069-9075.
  112. Oldfield BJ, Badoer E, Hards DK, McKinley MJ. 1994 Fos production in retrogradely labelled neurons of the lamina terminalis following intravenous infusion of either hyporetonic saline or angiotensin II. Neuroscience 60: 255-262.
  113. McKinley MJ, Burns P, Colvill LM, Wade JD, Weisinger RS, Tregear GW. 1997 Distribution of Fos immunoreactivity in the lamina terminalis and hypothalamus induced by centrally administered relaxin in consciuos rats. J. Neuroendocrinol. 9: 431-437.
  114. Farrell MJ, Bowala TK, Gavrilescu M, Phillips PA, McKinley MJ, McAllen RM, Denton DA, Egan GF.  2011 Cortical activation and lamina terminalis functional connectivity during thirst and drinking in humans. Am J Physiol Regul Integr Comp Physiol 301: R623–R631.
  115. Hoffman GE, Gibbs FP. 1982 Deafferentation spares a subchiasmatic LHRH projection to the median eminence. Neuroscience 8: 1979-1993.
  116. Ambach G, Palkovits M, Szentagothai J. 1976 Blood supply of the rat hypothalamus. IV. Retrochiasmatic area, median eminence, arcuate nucleus. Acta Morphol Acad Sci Hung 24: 93-119.
  117. Hoffman GE, Lee W-S, Wray S.Gonadotropin-releasing hormone (GnRH).In Nemeroff CB Ex., Neuroendocrinology, Boca Raton, FL, CRC Press, 1992, 185-217. 126.   Jansen HT, Kirbi JD, Cooke PS, Arambepola N, Iwamoto GA.  2007 Impact of neonatal hypothyroidism on reproduction in the male hamster, Mesocricetus auratus. Physiol Behav 90: 771-781.
  118. Toni R, Della Casa C, Castorina S, Cocchi D, Celotti F. 2005 Effects of hypothyroidism and endocrine disruptor-dependent non-thyroidal illness syndrome on the GnRH-gonadotroph axis of the adult male rat. J Endocrinol Invest 28 (suppl to no 11): 20-27.
  119. Dellmann HD. 1979 The subfornincal organ. Int Rev Cytol 58: 333-421.
  120. Dellmann HD. 1985 Fine structural organizatiojn of the subfornical organ. A concise review. Brain Res Bull 15: 71-78.
  121. Dellmann HD, Simpson JB. 1976 Regional differences in the morphology of the rat subfornincal organ. Brain Res 116: 389-400.
  122. Gross PM. 1992 Circumventricular ogan capillaries. Prog. Brain Res. 91: 219-233.
  123. Swanon LW, Mogenson GJ. 1981 Neural mechanisms for functional coupling of autonomic, endocrine and somatomotor responses in adaptative behavior. Brain Res. Reviews 3: 2-34.
  124. Tanaka J, Nojima K, Yamamuro Y, Saito H, Nomura M. 1993 Responses of subfornical organ neurons projecting to the paraventricular nucleus to hemorrhage. Brain Res 608: 141-144.
  125. Krause EG, Melhorn SJ, Davis JF, Scott KA, Ma LY, deKloet AD, Benoit SC, Woods SC, Sakai RR. 2008 AT1 receptors in the subfornical organ mediate the drinking and hypothalamic-pituitary-adrenal response to systemic isoproterenol. Endocrinology
  126. Lind RW. The subfornical organ: neural connections, in: Gross P. (ed), Circumventricular Organs and Body Fluids, vol 1, CRC Press, pp. 27-42.
  127. Bourque CW, Oliet SHR, Richard D. 1994 Osmoreceptors, osmoreception and osmoregulation. Frontiers in Neuroendocrinology 15: 231-274.
  128. Ferguson AV. 1992 Neurophysiological analysis of mechanisms for the subfornical organ and area postrema involvement in autonomic control. Prog. Brain Res. 91: 413-421.
  129. Toni R, Mosca S, Ruggeri F, Valmori A, Orlandi G, Toni G, Lechan RM, Vezzadini P. 1995 Effect of hypothyroidism on vasoactive intestinal polypeptide-immunoreactive neurons in the forebrain-neurohypophysial nuclei of the rat brain. Brain Res. 682: 101-115.
  130. Sakai K, Agassandian K, Morimoto S, Sinnayah P, Cassell MD, Davisson RL, Sgimund CD. 2007 Local production of angiotensin II in the subfornical organ causes elevated drinking. J Clin. Invest 117: 1088-1095.
  131. Kai A, Ono K, Kawano H, Honda E, Nakanishi O. Inenaga K. 2006 Galanin inhibits neuronal activity in the subfornical organ in rat slice preparation. Neuroscience 143: 769-777.
  132. Samson WK, White MM, Price C, Ferguson AV.2007 Obestatin acts in brain to inhibit thirst.  Am J Physiol Regul Integr Comp Physiol 292: R637-R643.
  133. Hajdu I, Szentirmai E, Obai Jr F, Krueger JM.2003 Different brain structures mediate drinking and sleep suppression elicited by the somatostatin analog octreotide in rats.  Brain Res 994: 115-123.
  134. Toni R, Mosca S, Lechan RM, Jackson IMD, Vezzadini P. 1994 Immunocytochemical distribution of the thyrotropin-releasing hormone (TRH) prohormone (ProTRH) and its terminal peptides in the rat subfornical organ. Eu J Endocrinoll 130 (suppl 2): P4.073, 251.
  135. Young CN, Morgan DA, Butler SD, Mark AL, Davisson RL. 2013 The brain subfornical organ mediates leptin-induced increases in renal sympathetic activity but not its metabolic effects.  Hypertension. 6:737-44.
  136. Gross PM. 1985 The subfornical organ as a model of neurohumoral integration. Brain Res. Bull. 15: 65-70
  137. Nauta SJH, Haymaker W. 1969 Hypothalamic nuclei and fiber connections, in Haymaker W, Anderson E, Nauta WJH (eds): The Hypothalamus. Charles C. Thomas, pp. 136-209
  138. Swanson LW. 1987 The hypothalamus, in Bjorklund A. Hokfelt T, Swanson LW (eds), Handbook of Chemical Neuroanatomy, vol 5, Integrated Systems of the CNS, part 1, Elsevier Science Publisher, pp 1-124.
  139. Vergani F, Landi A, Antonini A, Parolin M, Cilia R, Grimaldi M, Ferrarese C, Gaini SM, Sganzerla EP. 2007 Anatomical identification of active contacts in subthalamic deep brain stimulation.Mov Disord 18: 1337-1340.
  140. Novakova L, Ruzika E, Jech R, Serranova T, Dusek P, Urgosik D.2007 Increase in body weight in a non-motor side effect of deep brain stimulation of subthalamic nucleus in Parkinson’s disease.  Neuro Endocrinol Lett 28: 21-25.
  141. Szentagothai J. 1964 The parvicellular neurosecretory system, in Bargmann W, Shale JP (eds), Progress in Brain Research, vol 5, pp 135-146.
  142. Lechan RM, Alpert LC, Jackson IMD. 1976 Synthesis of luteinising hormone releasing factor and thyrotropin-releasing factor in glutamate-lesioned mice. Nature 264:463-465.
  143. Broadwell RD, Brightman MW. 1976 Entry of peroxidase into neurons of the central and peripheral nervous systems from extracerebral and cerebral blood. J Comp Neurol 166:257-284.
  144. Lechan RM, Nestler JL, Jacobson S, Reichlin S. 1980 The hypothalamic tuberoinfundibular system of the rat as demonstrated by horseradish peroxidase (HRP) microiontophoresis. Brain Research 195:13-27.
  145. Wiegand SJ, Price JL. 1980 The cells of the afferent fibers to the median eminence in the rat. J Comp Neurol 192:1-19.
  146. Lechan RM, Nestler JL, Jacobson S. 1982 The tuberoinfundibular system of the rat as demonstrated by immunohistochemical localization of retrogradely transported wheat germ agglutinin (WGA) from the median eminence. Brain Research 245:1-15.
  147. Dahlstrom A, Fuxe K. 1964 Evidence for the existence of monoamine-containing neurons in the central nervous system. I. Demonstration of monoamine in the cell bodies of brain stem neurons. Acta Physiol Scand, Suppl, 232:1-55.
  148. Niimi M, Takahara J, Sato M, Kawanishi K. 1989 Sites of origin of growth hormone-releasing factor-containing neurons projecting to the stalk-median eminence of the rat. Peptides 10:605-608.
  149. Niimi M, Takahara J, Sato M, Kawanishi K. 1992 Identification of dopamine and growth hormone-releasing factor-containing neurons projecting to the median eminence of the rat by combined retrograde tracing and immunohistochemistry. Neuroendocrinology 55:92-96.
  150. Merchenthaler I, Lennard DE. 1991 The hypophysiotropic neurotensin-immunoreactive neuronal system of the rat brain. Endocrinology 129:2875-2880.
  151. Niimi M, Takahara J, Sato M, Kawanishi K. 1991 Neurotensin and growth hormone-releasing factor-containing neurons projecting to the median eminence of the rat: A combined retrograde tracing and immunohistochemical study. Neurosci Letters 133:183-186.
  152. Takatsu Y, Matsumoto H, Ohtaki T, Kumano S, Kitada C, Onda H, Nishimura O, Fujino M. 2001 Distribution of galanin-like peptide in rat brain. Endocrinology 142: 1626-1634.
  153. Merchenthaler I. 1992 Enkephalin-immunoreactive neurons in the parvicellular subdivisions of the paraventricular nucleus project to the external zone of the median eminence. J Comp Neurol 326:112-120.
  154. Ishikawa K, Taniguchi Y, Kurosumi K, Suzuki M, Shinoda M. 1987 Immunohistochemical identification of somatostatin-containing neurons projecting to the median eminence of the rat. Endocrinology 121:94-97.
  155. Swanson LW, Kuypers HGJM. 1980 The paraventricular nucleus of the hypothalamus: Cytoarchitectonic subdivisions and organization of projections to the pituitary, dorsal vagal complex, and spinal cord as demonstrated by retrograde fluorescence double-labeling methods. J Comp Neurol 194:555-570.
  156. Kawano H, Tsuruo Y, Bando H, Daikoku S. Hypophysiotrophic TRH-producing neurons identified by combining immunohistochemistry for pro-TRH and retrograde tracing. 1991 J Comp Neurol 307:531-538.
  157. Niimi M, Takahara, Hashimoto K, Kawanishi K. 1988 Immunohistochemical identification of corticotropin releasing factor-containing neurons projecting to the stalk-median eminence of the rat. Peptides 9:589-593.
  158. Kawano H, Daikoku S, Shibasaki T. 1988 CRF-containing neuron systems in the rat hypothalamus: retrograde tracing and immunohistochemical studies. J Comp Neurol 272:260-268.
  159. Dalcik H, Phelps J. 1993 Median eminence-afferent vasoactive intestinal peptide (VIP) neurons in the hypothalamus: localization by simultaneous tract tracing and immunocytochemistry. Peptides 14:1059-1066.
  160. Merchenthaler I, Setalo G, Csontos C, Petrusz P, Flerko B, Negro-Vilar A. 1989 Combined retrograde tracing and immunocytochemical identification of luteinizing hormone-releasing hormone-and somatostatin-containing neurons projecting to the median eminence of the rat. Endocrinology 125:2812-2821.
  161. Kawano H, Tsuruo Y, Bando H, Daikoku S. 1991 Hypophysiotropic TRH-producing neurons identified by combining immunohistochemistry for pro-TRH and retrograde tracing. J Comp Neurol 307: 531-538.
  162. Fekete C, Mihaly E, Luo L-G, Kelly J, Clausen JT, Mao QF, Rand WM, Moss LG, Kuhar M, Emerson CH, Jackson IMD, Lechan RM. 2000 Association of cocaine- and amphetamine-regulated transcript-immunoreactive elements with thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and its role in the regulation of the hypothalamic-pituitary-thyroid axis during fasting. J Neurosci 20: 9224-9234.
  163. Silverman AJ, Jhamandas J, Renaud LP. 1987 Localization of luteinizing hormone-releasing hormone (LHRH) neurons that project to the median eminence. J Neurosci 7:2312-2319.
  164. Goldsmith PC, Thind KK, Song T. 1990 Location of the neuroendocrine gonadotropin-releasing hormone neurons in the monkey hypothalamus by retrograde tracing and immunostaining. J Neuroendocrinology 2:157-168.
  165. Rugarli EI, Ballabio A. 1993 Kallmann syndrome, from genetics to neurobioloogy. JAMA 270:2713-2716.
  166. Merchenthaler I. 1991 Neurons with access to the general circulation in the central nervous system of the rat: A retrograde tracing study with fluoro-gold. Neuroscience 44:655-662.
  167. Palkovits M, Leranth Cs, Zaborszky L, Brownstein MJ. 1977 Electron microscopic evidence of direct neuronal connections from the lower brain stem to the median eminence. Brain Res 136:339-344.
  168. Reeves WB, Andreoli TE. 1992 The posterior pituitary and water metabolism; Wilson, JD, Foster DW (eds); Williams Textbook of Endocrinology, W B Saunders Co, pp 311-356.
  169. Vandesande F, Dierickx K. 1975 Identification of vasopressin-producing and of the oxytocin-producing neurons in the hypothalamic magnocellular neurosecretory system of the rat. Cell Tissue Res 164:153-162.
  170. Stillman MA, Recht LD, Rosario SL, Seif SM, Robinson AG, Zimmerman EA. 1977 The effects of adrenalectomy and glucocorticoid replacement on vasopressin and vasopressin-neurophysin in the zona externa of the median eminence of the rat. Endocrinology 101:42-49.
  171. Whitnall HM, Smyth D, Gainer H. 1987 Vasopressin coexists in half of the corticotropin-releasing factor axons present in the external zone of the median eminence in normal rats. Neuroendocrinology 45:420-424.
  172. Raff H. 1993 Interactions between neurohypophysial hormones and the ACTH-adrenocortical axis. Ann N Y Acad Sci 689:411-425.
  173. Plotsky PM, Bruhn TO, Vale W. 1985 Hypophysiotropic regulation of adrenocorticotropin secretion in response to insulin-induced hypoglycemia. Endocrinology 117:323-329.
  174. Pow DV, Morris JF. 1989 Dendrites of hypothalamic magnocellular neurons release neurohypophysial peptides by exocytosis. Neuroscience 32:435-439
  175. Theodosis DT, Bonfanti L, Olive S, Rougon G, Poulain DA. 1994 Adhesion molecules and structural plasticity of the adult hypothalamo-neurohypophysial system. Psychoneuroendocrinology 19: 455-462.
  176. Theodosis DT. 2002 Oxytocin-secreting neurons: A physiological model of morphological neuronal and dglial plasticity in the adult hypothalamus. Front Neuroendo 23: 101-135
  177. Morris JF, Pow DV. 1993 New anatomical insights into the inputs and outputs from hypothalamic magnocellular neurons. Ann N Y Acad Sci 689:16-33.
  178. Chowdrey HS, Lightman SL. 1993 Role of central amino acids and peptide-mediated pathways in neurohypophysial hormone release. Ann N Y Acad Sci 689:183-193.
  179. Mikkelsen JD, Schmidt P, Sheikh SP, Larsen PJ. 1992 Non-vasopressinergic, non-oxytocinergic neuropeptides in the rat hypothalamo-neurohypophyseal tract: Experimental immunohistochemical studies. Progress in Brain Res 91:367-371.
  180. Nakamura S, Naruse M, Naruse K, Shioda S, Nakai Y, Uemura H. 1993 Colocalization of immunoreactive endothelin-1 and neurohypophysial hormones in the axons of the neural lobe of the rat pituitary. Endocrinology 32:530-533.
  181. Brown CH. 2016.Magnocellular neurons and posterior pituitary function, Compr Physiol 6: 1701-1741. 
  182. Stopa EG, LeBlanc VK, Hill DH, Anthony ELP. 1993 A general overview of the anatomy of the neurohypophysis. Ann N Y Acad Sci 689:6-15.
  183. Mohr E, Fehr S, Richter D. 1991 Axonal transport of neuropeptide encoding mRNAs within the hypothalamo-hypophyseal tract of rats. EMBO J 10:2419-2424.
  184. Skutella T, Probst JC, Blanco E, Jirikowski GF. 1994 Localization of tyrosine hydroxylase mRNA in the axons of the hypothalamo-neurohypophysial system. Mol Brain Res 23:179-184.
  185. Moos F, Richard P. 1982 Excitatory effect of dopamine on oxytocin and vasopressin reflex releases in the rat. Brain Res 241:249-260.
  186. Larsen PJ, Jukes KE, Chowdrey HS, Lightman SL, Jessop DS. 1994 Neuropeptide-Y potentiates the secretion of vasopressin from the neurointermediate lobe of the rat pituitary gland. Endocrinology 134:1635-1639.
  187. Larsen PJ, Sheikh SP, Mikkelsen JD. 1992 Osmotic regulation of neuropeptide Y and its binding sites in the magnocellular hypothalamo-neurohypophysial pathway. Brain Res 573:181-189.
  188. Gonzalez AM, Logan A, Ying W, Lappi DA, Berry M, Baird A. 1994 Fibroblast growth factor in the hypothalamic-pituitary axis: Differential expression of fibroblast growth factor-2 and a high affinity receptor. Endocrinology 134:2289-2297.
  189. Fagin KD, Wiener SG, Dallman MF. 1985 ACTH and corticosterone secretion in rats following removal of the neurointermediate lobe of the pituitary gland. Neuroendocrinology 40:352-362.
  190. Murai I, Ben-Jonathan N. 1987 Posterior pituitary lobectomy abolishes the suckling-induced rise in prolactin: Evidence for a prolactin-releasing factor in posterior pituitary. Endocrinology 121:205-211.
  191. Murai I, Ben-Jonathan N. 1990 Acute stimulation of prolactin release by estradiol: Mediation by the posterior pituitary. Endocrinology 126:3179-3184.
  192. Lechan RM, Arkun K, Toni R. 2019. Pituitary anatomy and development, in Prolactin Disorders, NA Tritos, A Klibanski, Eds, Humana Press: 11-53.
  193. Saeki N, Sunami K, Kubota M, Murai H, Takanashi J, Iuchi T, Yamaura A. 2001 Heavily T2-weighted MR imaging of white matter tracts in the hypothalamus: normal and pathologic demonstrations. AJNR 22: 1468-1475.
  194. Anderson E., Haymaker W. 1974 Breakthroughs in hypothalamic and pituitary research. Prog. Bran Res. 41: 1-60
  195. Kuhlenbeck H. 1973 The central nervous system of vertebrates. Overall morphologic pattern. Vol 3, Karger, p. 717.
  196. Haymaker W. 1969 Blood supply of the human hypothalamus, in Haymaker W, Anderson E, Nauta WJH (eds): The Hypothalamus. Charles C. Thomas, pp. 210-218.
  197. Page RB. 1986 The pituitary portal system, in Ganten D., Pfaff D. (eds), Morphology of the Hypothalamus and its connections, Springer-Verlag, pp 1-48.
  198. Szentagothai J. Flerko B, Mess B, Halasz B. 1972 Hypothalamic control of the anterior pituitary. An experimental-morphological study., 3 ed, Akadémiai Kiaidò, pp 87-92.
  199. Popa G., Fielding U. 1930 A portal circulation from the pituitary to the hypothalamic region. J. Anat. 65: 88-91.
  200. Fuxe K, Agnati LF, Harfstrand A, Cintra A, Aronsson M, Zoli M Gustafsson J-A. 1988 Principles for the hormone regulation of wiring transmission and volume transmission in the central nervous system, in Ganten D., Pfaff D. (eds), Neuroendocrinology of Mood, Springer-Verlag, pp 1-54.
  201. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M.2012A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid b. Sci Transl Med 4:147ra111.
  202. Rennels ML, Gregory TF, Blaumanis OR, Fujimoto K, Grady PA. 1985 Evidence for a ‘‘paravascular’’ fluid circulation in the mammalian central nervous system, provided by the rapid distribution of tracer protein throughout the brain from the subarachnoid space. Brain Res 326:47- 63.
  203. Jessen NA, Munk AS, Lundgaard I, Nedergaard M 2015 The Glymphatic system: A beginner's guide Neurochem Res. 40:2583-2599.
  204. Johnston M, Zakharov A, Papaiconomou C, Salmasi G, Armstrong D. 2004 Evidence of connections between cerebrospinal fluid and nasal lymphatic vessels in humans, non-human primates and other mammalian species. Cerebrospinal Fluid Res 1:2. doi:10.1186/ 1743-8454-1-2
  205. Jiang Q, Zhang L, Ding G, Davoodi-Bojd E, Li Q, Li L, Sadry N, Nedergaard M, Chopp M, Zhang Z2016  Impairment of the glymphaticsystem after diabetes. J Cereb Blood Flow Metab. 2016 pii: 0271678X16654702. [Epub ahead of print]
  206. Lechan R M, Fekete C, Toni R 2009 Thyroid Hormone in Neural Tissue. In: Donald W. Pfaff, Arthur P. Arnold, Anne M. Etgen, Susan E. Fahrbach and Robert T. Rubin (Eds), Hormones, Brain and Behavior, 2nd edition, Vol 2. San Diego: Academic Press; 2009. pp. 1289-1328.
  207. Agnati LF, Genedani S, Lenzi PL, Leo G, Mora F, Ferre S, Fuxe K 2005 Energy gradients for the homeostatic control of brain ECF composition and for VT signal migration: introduction of the tide hypothesis. J Neural Transm112:45–63
  208. Mascagni P.1787 De lymphaticis profundis capitis et colli In: Vasorum lymphaticorum corporis humani historia et ichonographia. Pars Prima Sectio VII, Art. VI, Siena: Pazzini Carli, pp. 63-64.
  209. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K. 2015 A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 212, 991–999
  210. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J.2015 Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341.

220  Bucchieri F, Farina F, Zummo G, Cappello F. 2015  Lymphatic vessels of the dura mater: a new discovery? J Anat. 227:702-703.

  1. Iliff JJ, Goldman SA, Nedergaard M. 2015 Implications of the discovery of brain lymphatic pathways. Lancet Neurol. 10:977-999.
  2. Cseer HF, Knopf PM 1992 Cervical lymphatics, the blood-brain barrier and the immunoreactivity of the brain: new view. Immunology Today 13:508-512.
  3. Lécuyer MA, Kebir H, Prat A. 2016 Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim Biophys Acta.1862:472-82.
  4. Dyess EM, Segerson TP, Liposits Z, Paull WK, Kaplan MM, WU P. Jackson IMD, Lechan RM. 1988 Triiodothyronine exerts direct cell-specific regulation of thyrotropin-releasing hormone gene expression in the hypothalamic paraventricular nucleus. Endocrinology123:2291-2297.
  5. Lechan R.M., Kakucska I. 1992 Feedback regulation of thyrotropin-releasing hormone gene expression by thyroid hormone in the hypothalamic paraventricular nucleus, Functional Anatomy of the Neuroendocrine Hypothalamus, Wiley, Chichester, Ciba Foundation Symposium 168, pp 144-164.
  6. Tannenbaum G.S., Martin JB. 1976 Evidence for an endogenous ultradian rhythm governing growth hormone secretion in the rat. Endocrinology 1976; 98:2255-2257.
  7. Yamauchi N, Shibasaki T, Ling N, Demura H. In vitro release of growth hormone-releasing factor (GRF) from the hypothalamus: somatostatin inhibits GRH release. Regul Pept 1991, 33: 71-78;de los Frailes MT, Cacicedo L, Fernandez G, et al, Role of locally produced growth-hormone-releasing factor in somatostatin regulation by fetal rat brain cells in culture, Neuroendocrinology 1992, 55: 221-229 ).
  8. Sato M, Frohman LA. Differential effects of central and peripheral administration of growth hormone (GH) and insulin-like growth factor on hypothalamic GH-releasing hormone and somatostatin gene expression in GH-deficiency dward rats, Endocrinology 1993, 133: 793-7990
  9. Epelbaum J. 1992 Intrahypothalamic neurohormonal interactions in the control of growth hormone secretion. Functional Anatomy of the Neuroendocrine Hypothalamus, Wiley, Chichester, Ciba Foundation Symposium 168, pp 54-68.
  10. Minami S, Kamegai J, Sugihara H et al. Growth hormone inhibits its own secretion by acting on the hypothalamus through its receptors on neuropeptide Y neurons in the arcuate nucleus and somatostatin neurons in the periventricular nucleus. Endocr J, 1998, 45(suppl): S19-S26)
  11. Casanueva FF, Villanueva L, Cabranes JA, Cabezas-Cerrato J, Fernandez-Cruz A. 1984 Cholinergic mediation of growth hormone Endocrinol Metab 59: 526-530.
  12. Cuttler L. 1996 The regulation of growth hormone secretion, End and Metab Clin. NA 25: 541-571.
  13. Nemeroff CB, Mason GA, Bissette G, Parks R, Schwarcz DA. 1985 Effects of intra-hypothalamic-injection of quinolinic acid on anterior pituitary hormone secretion in the unanesthetized rat. Neuroendocrinology 41: 332-337.
  14. Acs Z, Lonart G, Makara GB. 1990 Role of hypothalamic factors (growth-hormone-releasing hormone and gamma-aminobutyric acid) in the regulation of growth hormone secretion in the neonatal and adult rat. J. Neuroendocrinol. 52: 156-160.
  15. Kiss J, Csaba Z, Csaki A. Halasz B. 2006 Glutamatergic innervation of growth hormone-releasing hormone-containing neurons in the hypothalamic arcuate nucleus and somatostatin-containing neurons in the anterior periventricular nucleus of the rat. Brain Res Bull 70: 278-288.
  16. Radcliff RP, Lookingland KJ, Chapin LT, Tucker HA. 2001 Pituitary adenylate cyclase-activating polypeptide induces secretion of growth hormone in cattle. Domest Anim Endocrinol 2: 187-196.
  17. Adams BA, Gray SL, Isaac ER, Bianco AC, Vidal-Puig AJ, Sherwood NM. 2008 Feeding and metabolism in mice lacking pituitary adenylate cycolase-activating polypeptide. Endocrinology 149: 1571-1580.
  18. Anderson LL, Scanes CG. 2012 Nanobiology and physiology of growth hormone secretion. , Exp Biol Med 237: 126-142.
  19. Smith RG, Van der Ploeg LH, Howard AD, Feighner SD, Cheng K, Hickey GJ, Wyvratt MJ Jr, Fisher MH, Nargund RP, Patchett AA. 1997 Peptidomimetic regulation of growth hormone secretion. Endocr Rev 18: 621-645.
  20. Kojima H, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. 1999 Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402: 656-660.
  21. Muller et al. Gherelin. 2015 Mol Metab, 4: 437-460.
  22. Muccioli G, Tschop M, Tapotti M, Deghenghi R, Heiman M, Ghigo E. 2002 Neuroendocrine and peripheral activities of ghrelin: implications in metabolism and obesity. Eu J Pharmacol 440: 235-254.
  23. Grouselle D, Chaillou E, Caraty A, Bluet-Pajot MT, Zizzari P, Tillet Y, Epelbaum J. 2008 Pulsatile cerebrospinal fluid and plasma ghrelin in relation to growth hormone secretion and food intake in the sheep.J Neuroendocrinol 20: 1138-1146.
  24. Tschop M, Flora DB, Mayer JP, Heiman ML. 2002 Hypophysectomy prevents sghrelin-induced adiposity and increases gastric ghrelin secretion in rats. Obesity Res 10: 991-999.
  25. Steyn FJ, Tolle V, Chen C, Epelbaum J. Neuroendocrine regulation of growth hormone ssecretion. Compr Physiol 2016 6: 687-735.)
  26. Hillard CJ, Beatka M, Sarvaido J. Endocannabinoid signaling and the hypoyhalamic-pituitary-adrenal axis, Compr Physiol 2016, 7:1-15.
  27. Sawchenko PE, Swanson LW. 1981 Central noradrenergic pathways for the integration of hypothalamic neuroendocrine and autonomic responses. Science 214:685-687.
  28. Sawchenko PE, Swanson LW, Grzanna R, Howe PRC, Polack J, Bloom SR. 1985 Co-localization of neuropeptide Y-immunoreactivity in brainstem catecholaminergic neurons that project to the paraventricular nucleus of the hypothalamus. J Comp Neurol 249. Plotsky PM, Kjaer A, Sutton SW, Sawchenko PE, Vale W. 1991 Central activin administration modulates corticotropin-releasing hormone and adrenocorticotropin secretion. Endocrinology128: 2520-2525.
  29. Sarkar S, Fekete C, Legradi G, Lechan RM. 2003 Glucagon like peptide-1(7-36) amide (GLP-1) nerve terminals densely innervate corticotropin-releasing hormone neurons in the hypothalamic paraventricular nucleus. Brain Res 985: 163-168
  30. Liposits Zs, Sherman D, Phelix C, Paull WK. 1986 Adrenergic innervation of corticotropin releasing factor (CRF)-synthesizing neurons in the hypothalamic paraventricular nucleus in the rat. Histochemistry 84:201-205.
  31. Liposits Zs, Sievers L, Paull WK. 1988 Neuropeptide-Y and ACTH-immunoreactive innervation of corticotropin releasing factor (CRF)-synthesizing neurons in the hypothalamus of the rat. Histochemistry 88:227-234.
  32. Saper CB, Lowey AD, Swanson LW, Cowan WM. 1976 Direct hypothalamo-autonomic connections. Brain Res 117: 305-312.
  33. Sawchenko PE, Li H-Y, Ericsson A. 2000 Circuits and mechanisms governing hypothalamic responses to stress: a tale of two paradigms. Progress in Brain Res 122: 61-77.
  34. Besedovsky HO, Del Rey A. 1996 Immune-neuro-endocrine interactions: facts and hypotheses. Endocr Rev 17: 64-102.
  35. Ericsson A, Kovacs K, Sawchenko PE. 1994 A functional anatomical analysis of central pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons. J Neurosci 14: 897-913.
  36. Rivest S, Lacroix S, Vallieres L, Nadeau S, Zhang J, Laflamme N. 2000 How the blood talks to the brain parenchyma and the paraventricular nucleus of the hypothalamus during systemic inflammatory and infectious stimuli. Proc Soc Exp Biol Med 223: 22-28.
  37. Ericsson A, Arias C, Sawchenko PE. 1997 Evidence for an intramedullary prostaglandin-dependent mechanism in the activtion of stress-related neuroendocrine circuitry by intravenous interleukin-1. J Neurosci 17: 7166-7179.
  38. Li H-Y, Ericsson A, Sawchenko PE. 1996 Distinct mechanisms underlie activation of hypothalamic neurosecretory neurons and their medullary catecholaminergic afferents in categorically different stress paradigms. Proc Natl Acad Sci, USA 93: 2359-2364.
  39. Herman JP, Cullinan WE, Young EA, Akil H, Watson SJ. 1992 Selective forebrain fiber tract lesions implicate ventral hippocampal structures in tonic regulation of paraventricular nucleus corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP) mRNA expression. Brain Res 592:228-238.
  40. Herman JP, Cullinan WE, Watson SJ. 1994 Involvement of the bed nucleus of the stria terminalis in tonic regulation of paraventricular hypothalamic CRH and AVP mRNA expression. J Neuroendocrinology 6:433-442.
  41. Tuchelt H, Dekker K, Bahr V, Oelkers W. 2000 Dose-response relationship between plasma ACTH and serum cortisol in the insulin-hypoglycaemia test in 25 healthy subjects and 109 patients with pituitary disease, Clin Endocrinol 53: 301-307.
  42. Dimitrov EL, DeJoseph MR, Brownfield MS, Urban JH. 2007 Involvement of neuropeptide YY1 receptors in the regulation of neuroendocrine corticotropin-releasing hormone neuronal activity. Endocrinology 148: 3666-3673.
  43. Levy BH, Tasker JG. 2012 Synaptic regulation of the hypothalamic-pituitary-adrenal axis and its modulation by glucocorticoids and stress. Front Cell Neurosci 6: 1-13.
  44. Singru PS, Sanchez E, Acharya R, Fekete C, Lechan RM. 2008 Mitogen-activated protein kinase contributes to lipopolysaccharide-induced activation of corticotropin-releasing hormone synthesizing neurons in the hypothalamic paraventricular nucleus. Endocrinology 149: 2283-2292.
  45. Khan AM, Watts AG 2004 Intravenous 2-deoxy-D-glucose injection rapidly elevates levels of the phosphorylated forms of p44/42 mitogen-activated protein kinases (extracellularly regulated kinases 1/2) in rat hypothalamic parvicellular paraventricular neurons. Endocrinology 145:351-359.
  46. Sasaguri K, Kikuchi M, Hori N, Yuyama N, Onozuka M, Sato S. 2005 Suppression of stress immobilization-induced phosphorylation of ERK 1/2 by biting in the rat hypothalamic paraventricular nucleus. Neurosci Lett 383:160-164
  47. Abel ED, Ahima RS, Boets ME et al. 2001. Critical role for thyroid hormone receptor beta 2 in the regulation of paraventricular thyrotropin-releaing hormone n;eurons. J Clin Invest 107:1017-1023.
  48. Lechan RM, Kakucska I. 1992 Feedbsck regulation of thyrotropin-releasing hormone gene expressionby thyroid hormone in the hypothalamic paraventricular nucleus. Ciba Found Symp 168: 144-158; discussion 158-164.
  49. Fekete C, Lechan RM.2014. Central regulation of hypothalamic-pituitary-thyroid axis under physiological and pathophysiological conditions. Endocr Rev 35:159-194.
  50. Farkas, E., E. Varga, B. Kovacs, A. Szilvasy-Szabo, A. Cote-Velez, Z. Peterfi, M. Matziari, M. Toth, D. Zelena, Z. Mezricky, A. Kadar, D. Kovari, M. Watanabe, M. Kano, K. Mackie, B. Rozsa, Y. Ruska, B. Toth, Z. Mate, F. Erdelvi, G. Szabo, B. Gereben, R. M. Lechan, J. L. Charli, P. Joseph-Bravo and C. Fekete. 2020. A glial-neuronal circuit in the median eminence regulates thyrotropin-releasing hormone-release via the endocannabinoid system. iScience 23(3): 100921.
  51. Sanchez E, Vargas MA, Singru PS, Pascual I, Romero F, Fekete C, Charli JL, Lechan RM. 2009. Tanycyte pyroglutamyl peptidase II contributes to regulation of the hypothalamic-pituitary-thyroid axis through glial-axonal associations in the median eminence. Endocrinology 150: 2283-2291.
  52. Blake NG, Eckland JA, Foster OJF, Lightman SL. 1991 Inhibition of hypothalamic thyrotropin-releasing hormone messenger ribonucleic acid during food deprivation. Endocrinology 129: 2714-2718,
  53. Legradi G, Emerson CH, Ahima RS, Flier JS, Lechan RM. 1997 Leptin prevents fasting-induced suppression of prothyrotropin-releasing hormone messenger ribonucleic acid in neurons of the hypothalamic paraventricular nucleus. Endocrinology 138: 2569-2576.
  54. Rondeel JMM, Heide R, deGreef WJ, van Toor H, van Haasteren GAC, Klootwijk W, Visser TH. 1992 Effect of starvation and subsequent refeeding on thyroid function and release of hypothalamic thyrotropin-releasing hormone. Neuroendocrinology 56: 348-353.
  55. Harris RC, Fang S-L, Azizi F, Lipworth L, Vagenakis AG, Braverman LE. 1978 Effect of starvation on hypothalamic-pituitary-thyroid function in the rat. Metabolism 27: 1074-1083.
  56. Flier JS, Harris M, Hollenberg AN. 2000 Leptin, nutrition, and the thyroid: the why, the wherefore, and the siring. J Clin Invest 105: 859-861.
  57. Fekete C, Legradi G, Mihaly E, Huang Q-H, Tatro JB, Rand WM, Emerson CH, Lechan RM. 2000 α-Melanocyte stimulating hormone is contained in nerve terminals innervating thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and prevents fasting induced suppression of prothyrotropin-releasing hormone gene expression. J Neurosci 20: 1550-1558.
  58. Fekete C, Kelly J, Mihaly E, Sarkar S, Rand WM, Legradi G, Emerson CH, Lechan RM. 2001 Neuropeptide Y has a central inhibitory action on the hypothalamic-pituitary-thyroid axis. Endocrinology 142: 2606-2613.
  59. Fekete C, Sarkar S, Rand WM, Harney JW, Emerson CH, Bianco AC, Lechan RM. 2002 Agouti related protein (AGRP) has a central inhibitory action on the hypothalamic-pituitary-thyroid (HPT) axis; comparisons between the effect of AGRP and neuropeptide Y (NPY) on energy homeostasis and the HPT axis. Endocrinology 143: 3846-3853.
  60. Toni R, Jackson IMD, Lechan RM. 1990 Neuropeptide Y-immunoreactive innervation of thyrotropin-releasing hormone-synthesizing neurons in the rat hypothalamic paraventricular nucleus. Endocrinology126:2444-2453.

282 Legradi G, Lechan RM. 1998 The arcuate nucleus is the major source for neuropeptide Y-innervation of thyrotropin-releasing hormone neurons in the hypothalamic paraventricular nucleus. Endocrinology 139: 3262-3270.

  1. Legradi G, Lechan RM. 1999 Agouti-related protein containing nerve terminals innervate thyrotropin-releasing hormoneneurons in the hypothalamic paraventricular nucleus. Endocrinology 140: 3643-3652.
  2. Fekete C, Sarkar S, Rand WM, Harney JW, Emerson CH, Bianco AC, Beck-Sickinger A, Lechan RM. 2002 Neuropeptide Y1 and Y5 receptors mediate the effects of NPY on the hypothalamic-ppituitary-thyroid (HPT) axis. Endocrinology 143: 4513-4519.
  3. Ollmann MM, Wilson BD, Yang YK, Kerns JA, Barsh GS, Kent SBH, Gantz I, Barsh GF. 1997 Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein, Science 278: 135-138.
  4. Ahima RS, Saper CB, Flier JS, Elmquist JK. 2000 Lepin regulation of neuroendocrine systems, Front Neuroendocrinology 21: 263-307.
  5. De Groot. 1999 Dangerous dogmas in medicine: the nonthyroidal illness syndrome. J Clin Endocrinol Metab 84: 151-164.
  6. Sergeyev V, Broberger C, Hokfelt T, 2001 Effect of LPS administration on the expression of POMC, NPY, galanin, CART and MCH mRNAs in the rat hypothalamus, Mol Brain Res 90: 93-100.
  7. Francis J, MohanKumar PS, MohanKumarSM, Quandri SK. 1999 Systemic administration of lipopolysaccharide increases plasma leptin levels: blockade by soluble interleukin-1 receptor. Endocrine 10: 291-295.
  8. Tapia-Arancibia L. Arancibia S, Astier H. 1985 Evidence for a1-adrenergic stimulatory control of in vitro release of immunoreactive thyrotropin-releasing hormone from rat median eminence: In vivo corroboration. Endocrinology 116: 2314-2319.
  9. Lechan RM, and Fekete C. 2007 Infundibular tanycytes as modulators of neuroendocrine function: hypothetical role in the regulation of the thyroid and gonadal axis. Acta Biomed 78 Suppl 1: 84-98.
  10. Heuer H, Maier MK, Iden S, Mittag J, Friesema EC, Visser TJ, and Bauer K. 2005 The monocarboxylate transporter 8 linked to human psychomotor retardation is highly expressed in thyroid hormone-sensitive neuron populations. Endocrinology 146: 1701-1606.
  11. Agnati LF, Zoli, Stromberg MI, and Fuxe K. 1995 Intercellular communication in the brain: wiring versus volume transmission. Neuroscience 69: 711-276.
  12. Diano S, Naftolin F, Goglia F, Csernus V, and Horvath TL. 1998 Monosynaptic pathway between the arcuate nucleus expressing glial type II iodothyronine 5'-deiodinase mRNA and the median eminence-projective TRH cells of the rat paraventricular nucleus. J Neuroendocrinol 10: 731-472.
  13. Freitas BC, Gereben B, Castillo M, Kallo I, Zeold A, Eqri P, Liposits Z, Zavacki AM, Maciel RM, Jo S, Singru P, Sanchez E, Lechan RM, Bianco AC. 2010 Paracrine signaling by glial cell-derived triiodothyronine activates neuronal gene expression in the rodent brain and human cells. J Clin Invest 120: 2206-22217.
  14. Joseph-Bravo P, Jaimes-Hoy L, Uribe R-M, Charli J-L. 2015. 60 years of neuroendocrinology: TRH, the first hypophysiotropic releasing hormone isolated: control of the pituitary-thyroid axis, J Endocrinol 226(2): T85-T100.
  15. Clark IJ, Cummigs JT. 1982 The temporal relationship between gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in ovariectomized ewes. Endocrinology 111: 1737-1739.
  16. Moenter SM, Caraty A, Locatelli A, Karsch FJ. 1991 Pattern of gonadotropin-releasing hormone (GnRH) secretion leading up to ovulation in the ewe: existence of a preovulatory GnRH surge. Endocrinology 129: 1175-1182.
  17. Johnson ES, Gendrich RL, White WF. 1976 Delay of puberty and inhibition of reproductive processes in the rat by a gonadotropin-releasing hormone agonist analog. Fertil. Steril.27: 853-860.
  18. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS J., Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O’Rahily S, Carlton MB, Crowley Jr WF, Aparicio SA, Colledge WH.2003 The GPR54 gene as a regulator of puberty.  N Eng J Med. 349: 1614-1627.
  19. Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ., Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P.2001    AXOR12, a novel human G protein-coupled receptor, activated by the peptide Kiss-1.  J. Biol. Chem 276: 28969-28975.
  20. Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E. 2001 The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem 276: 34631-34636.
  21. Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y.2001  Metastin suppresses the motility and growth of CHO cells transfected with its receptor. Nature 411: 613-617.
  22. King JC, Tobet SA, Snavley FL, Arimura AA. 1982 LHRH immunopositive cells and their projections to the median eminence and organum vasculosum of the lamina terminalis. J Comp Neurol 209(3):287-300.
  23. Vazquez-Martinez R, Shorte SL, Boockfor FR, Frawley LS. 2001 Synthronized exocytotic bursts from gonadotropin-releasing hormone-expressing cells: dual control by intrinsic cellular pulsatility and gap junctional commnication. 2001 Endocrinology 142: 2095-2101.
  24. Nunemaker CS, DeFazio RA, Geusz M., Herzog ED, Pitts GR, Moenter SM. 2001 Long-term recordings of networks of immortalized GnRH neurons reveal episodic patterns of electrical activity. 2001 J. Neurophysiol. 86: 86-93.
  25. Messager S, Chatzidaki EE, Ma D Hendrick AG. Zahn D, Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MB, Colledge WH, Caraty A, Aparicio SAJR. 2005 Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. PNAS 102: 1761-1766.
  26. Gottsch ML, Cunningham MJ, Smith JT, Popa SM, Acohido BV, Crowley WF, Seminara S, Clifton DK, Steiner RA. 2004Endocrinology 145: 4073-4077.
  27. Matsui H, Takatsu Y, Kumano S, Matsumoto H, Ohtaki T. 2004Peripheral administration of metastin induces marked gonadotropin release and ovulation in the rat.   Biochem. Biophys Res Commun 320: 383-388.
  28. Navarro VM, Castellano JM, Fernandez-Fernandez R, Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E, Dieguez CF, Pinilla L, Tena-Sempere M. 2004Developmental and hormonally regulated messenger ribonucleic acid expression of KISS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KISS-1 peptide. Endocrinology 145: 4565-4574.
  29. Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich SK, Clifton DK, Steiner RA, Herbison AE. 2005 Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci 25: 11349-11356.

312  D’Anglemont de Tassigny X, Fagg LA, Dixon JPC, Day K, Leitch HG, Hendrick AG, Zahn D, Franceschini I, Caraty A, Carlton MBL, Aparicio SAJR, Colledge WH. 2006 Hypogonadotropic hypogonadism in mice lacking a functional Kiss1 gene. PNAS 104: 10714-10719.

  1. D’Anglemont de Tassigny X, Fagg LA, Carlton MBL, Colledge WH. 2008 Kisspeptin can stimulate gonadotropin-releasing hormone (GnRH) release by a direct action at GnRH nerve termnals. Endocrinology 149: 3926-2932.
  2. Seminara SB, Dipietro MJ, Ramaswamy S, Crowley Jr WF, Plant TM. 2006 Continuous human metastin 45-54 infusion desensitizes G protein-coupled receptor 54-induced gonadotropin-releasinghormone release monitored indirectly in the juvenile male rhesus monkey (Macaca mulatta): a finding with therapeutic implications. Endocrinology 147: 2122-2126.
  3. Ramaswamy S, Guerriero KA, Gibbs RB, Plant TM. 2008 Structural interactions between kisspeptin and GnRH neurons in the mediobasal hypothalamus of the male Rhesus monkey (Macaca mulatta) as revealed by double immunofluorescence and confocal microscopy. Endocrinology 149: 4387-4395.
  4. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA. 2005 Regulation of Kiss 1 gene expression in the brain of the female mouse. Endocrinology 146: 3686-3692.
  5. Decourt C, Tillet Y, Caraty A, Franceschini I, Briant C. 2008 Kisspeptin immunoreactive neurons in the equine hypothalamus.Interactions with GnRH neuronal system, J. Chem Neuroanat 36:131-137.
  6. Franceschini I, Lomet D, Cateau M. Delsol G, Tillet Y. Caraty A. 2006 Kisspeptin immunoreactive cells of the ovine preoptic area and arcuate nucleus co-express estrogen receptor alpha. Neuroscience Letters 410: 225-230.
  7. Dungan HM, Clifton DK, Steiner RA. 2006 Kisspeptin neurons as central processors in the regulation of gonadotropin-releasing hormone secretion. Endocrinology 147: 1154-1158.
  8. Stephens SB, Tolson KP, Rouse MLI, Poling MC, Hashimoto-Parktya M, Mellon PL, Kauffman AS. 2015. Absent progesterone signaling in kisspeptin neurons disrupts the LH surge and impairs fertility in female mice. Endocrinology 156: 3091-3097.
  9. Kauffman AS, Gottsch ML, Roa J, Byquist AC, Crown A, Clifton DK, Hoffman GE, Steiner RA, Tena-Sempere M. 2007 Sexual differentiation of Kiss1 gene expression in the brainof the rat. Endocrinology 148: 1774-1783.
  10. Homma T, Sakakibara M, Yamada S, Kinoshita M, Iwata K, Tomikawa J, Kanazawa T, Matsui H, Takatsu Y, Ohtaki T, Matsumoto H, Uenoyama Y, Maeda K, Tsukamura H. 2009 Significance of neonatal testicular sex steroids to defeminize anteroventral periventricular kisspeptin neurons and the GnRH/LH surge system in male rats. Biol Reprod 81: 1216-1225.
  11. Lehman MN, Coolen LM, Goodman RL. 2010 Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology 151: 3479-3489.
  12. Roa J, Navarro VM, Tena-Sempere M. 2011 Kisspeptins in reproductive biology: consensus knowledge and recent developments. Biol Reprod 85: 650-660.
  13. Goodman RL, Hileman SM, Nestor CC et al. 2013. Kisspeptin, neurokinin B, and dynorphin act in the arcuate nucleus to control activity of the GnRH pulse generator in ewes. Endocrinology 154: 4259-4269.
  14. Treen AK, Luo V, Belsham DD. 2016 Phoenixin activates immortalized GnRH and kisspeptin neurons through the novel receptor GPR173. Mol Endocrinol 30: 872-888.
  15. Quennell JH, Howell CS, Roa J, Augustine RA, Grattan DR, Anderson GM. 2011 Leptin deficiency and diet-induced obesity reduce hypothalamic kisspeptin expression in mice. Endocrinology 152: 1541-1550.
  16. Ojeda SR, Lomniczi A, Sandau US. 2008 Glial-gonadotrophin hormone (GnRH) neurone interactions in the median eminence and the control of GnRH secretion, J. Neuroendocrinology 20: 732-742.
  17. Garcia-Segura M, Lorenz B, DonCarols LL. 2008 The role of glia in the hypothalamus: implications for gonadal steroid feedback and reproductive neuroendocrine output. Reproduction 135: 419-429.
  18. Prevot V., Dehouck B., Poulain P., Beauvillain J-C., Buee-Scherrer V., Bouret S. 2007 Neuronal-glial-endothelial interactions and cell plasticity in the postnatal hypothalamus: implications for the neuroendocrine control of reproduction. Psychoneuroendocrinology 32: S46-S51.
  19. Sharif A, Baroncini M, Prevot V. 2013. Role of glia in the regulation of gonadotropin-releasing hormone neuronal activity and secretion. Neuroendocrinology 98: 1-15.
  20. Langub M.C., Watson RE. Jr. 1992 Estrogen receptor-immunoreactive glia, endothelia, and ependyma in guinea pig preoptic area and median eminence: electron microscopy. Endocrinology 130: 364-372.
  21. Rivier C, Rivier J, Vale W. 1986 Stress-induced inhibition of reproductive functions: role of endogenous corticotropin-releasing factor, Science 231: 607-609.
  22. Freeman ME, Kanyicska B, Lerant A, Nagy G. 2000 Prolactin: structure, function and regulation of secretion. Physiol Rev 80: 1523-1631.
  23. Stricker EM, Verbalis JG. Water intake and body fluids, in Fundamental Neuroscience, ch 42, pp 1111-1126, 1999.
  24. Dunn FL, Brennan RJ, Nelson AE, Robertson GL. 1973 The role of blood osmolality and volume in regulatig vasopressin secretion in the rat. J Clin Invest 52: 3212-3219.
  25. Shaw S, Marples D. 2002 A rat kidney tubule suspension for the study of vasopressin-induced shuttling of AQP2 water channels. Am J Physiol Renal Physiol. 283: F1160-F1166
  26. Valenti G, Procino G, Tamma G, Carmosino M, Svelto M. 2005 Minireview: aquaporin 2 trafficking. Endocrinology 146: 5963-5070.
  27. Osaka T, Kannan H, Kasai M, Inenaga K, Yamashita H. 1990 Osmotic responses of rat paraventricular neurons by pressure ejection method. Brain Res. Bull. 24: 493-497.
  28. Honda K, Negoro H, Higuchi T, Tadokoro Y. 1987 Activation of neurosecretory cells by osmotic stimulation of anteroventral third ventricle. Am J Physiol. 252: R1039-1045.
  29. McKinley MJ, Bicknell RJ, Hards D, McAllen RM, Vivas L, Weisinger RS, Oldfield BJ. 1992 Efferent neural pathways of the lamina terminalis subserving osmoregulation. Prog. Brain Res. 91: 395-402.
  30. Ferguson AV, Renaud LP. 1986 Systemic angiotensin acts at subfornical organ to facilitate activity of neurohypophysial neurons. Am J Physiol 251: R712-R717.
  31. Liedtke W. 2007 Hydromineral neuroendocrinology: role of TRPV ion channels in sensory transduction of osmotic stimuli in mammals. Exp. Physiol. 92: 507-512.
  32. Forsling ML, Ingram DL, Stanier MW. 1976 Effects of various ambient temperatures and of heating and cooling the hypothalamus and cervical spinal cord on antidiuretic hormone secretion and urinary osmolality in pigs. J Physiol 257: 673-686.
  33. Azahan E, Sykes AH. 1980 The effects of ambient temperature on urinary flow and composition in the fowl. J Physiol 304: 389-396.
  34. Sharif-Naeini R, Ciura S, Bourque CW. 2008 TRPV1 gene required for thermosensory transduction and anticipatory secretion from vasopressin neurons during hyperthermia. Neuron 58: 179-185.
  35. Hiyama TY, Matsuda S, Fujikawa A, Matsumoto M, Watanabe E, Kajiwara H, Nimura F, Noda M. 2010 Autoimmunity to the sodium-level sensor in the brain causes essential hypernatremia. Neuron 66: 508-522.
  36. Dohanics J, Linton EA, Lowry PJ, Makara GB. 1990 Osmotic stimulation affects neurohypophysial corticotropin releasing factor-41 content: Effect of dexamethasone. Peptides 11:51-57.
  37. Oelkers W. 1989 Hyponatremia and inappropriate secretion of vasopressin (antidiuretic hormone) in patients with hypopituitarism. N Engl J Med 321:492-496.
  38. Kiss JZ, Van Eekelen AM, Reul JMHM, Westphal HM, DeKloet ER. 1985 Glucocorticoid receptor in magnocellular neurosecretory cells. Endocrinology 122:444-449.
  39. Morris JF, Pow DV. 1993 New anatomical insights into the inputs and outputs from hypothalalmic magnocellular neurons. Ann N Y Acad Sci 689: 16-33.
  40. Chowdrey HS, Lightman SL. 1993 Role of central amino acids and peptide-mediated pathways in neurohypophysial hormone release. Ann N Y Acad Sci 689: 183-193.
  41. Hetherington AW, Ranson SW. 1940 Hypothalamic lesions and adiposity in the rat, Anat Rec 78: 149-172
  42. Reeves AG and Plum F. 1969 Hyperphagia, rage amd dementia accompanying a ventromedial hypothalamic neoplasm. Arch Neurol 20: 616-624.
  43. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. 1994 Positional cloning of the mouse obese gene and its human homologue, Nature 372: 425-432.
  44. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. 2000 Central nervous system control of food intake, Nature 404: 661-671.
  45. Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS. 1999 Interacting appetite-regulating pathways in the hypothalamic regulation of body weight, Endocr Rev 20: 68-100.
  46. Barsh GS, Schwartz. 2002 Genetic approaches to studying energy balance: perception and integration, Nature Rev 3: 589-600.
  47. Gautren L, Elmquist JK. 2011 Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 121: 2087-2093.
  48. Zhao AZ, Huan J-N, Gupta S, Pal R, Sahu A. 2002 A phosphatidylinositol 3-kinase phosphodiesterase 3B-cyclic AMP pathway in hypothalamic action of leptin on feeding. Nat. Neurosci. 5: 727-728.
  49. Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, Neel BG, Schwartz MW, Myers Jr MG. 2003 STAT3 signaling is required for leptin regulation of energy balance but not reproduction. Nature 421: 856-859.
  50. Kwon O, Kim KW, Kim M-S. 2016 Leptin signaling pathways in hypothalamic neurons, Cell Mol Life Sci 73: 1457-1477.
  51. Nejenhuis WAJ, Oosterom J, Adan RAH. 2001 AgRP (83-132) acts as an inverse agonist on the human-melanocortin-4 receptor, Mol Endocrinol 15: 164-171.
  52. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL, Cone RD, Low MJ. 2001 Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus, Nature 411: 480-484.
  53. Erickson JC, Clegg KE, Palmiter RD. 1996 Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y, Nature 381: 415-421.
  54. Huzar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F. 1997 Targeted disruption of the melanocortin-4 receptor results in obesity in mice, Cell 88: 131-141.
  55. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. 1998 Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans, Nat Genet 19: 1155-1157.
  56. Cheetham T, O’Rahilly S. 2000 Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency, J Clin Invest 106: 271-279.
  57. Jackson RS, Creemers JW, Ohagi S, Raffin-Sanson ML, Sanders L, Montague CT, Hutton JC, O’Rahilly S. 1997 Obesisty and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene, Nature Genet 6: 218-220.
  58. Gruenewald DA, Matsumoto AM. 1991 Age-related decrease in proopiomelanocortin gene expression in the arcuate nucleus of the male rat brain, Neurobiol Aging 12: 113-121.
  59. Mobbs CV, Bray GA, Atkinson RL, Bartke A, Finach CE, Maratos-Flier E, Crawley JN. Nelson JF. 2001 Neuroendocrine and pharmacological manipulations to assess how caloric restriction increase lifespan, J Gerontology 56: 34-44.
  60. Wisse BE, Frayo RS, Schwartz MW, Cummings DE. 2001 Reversal of cancer anorexia by blockade of central melanocortin receptors in rats, Endocrinology 142: 3292-3101.
  61. Marks DL, Ling N, Cone RD. 2001 Role of the central melanocortin system in cachexia, Cancer Res 61: 1432-1438.
  62. Kuhnen K, Clement S, Wiegand et al. 2018 Proopiomelanocortin deficiency treated with a melanocortih-4 receptor agonist. N Engl J Med 2016; 375: 240-246, Clement H Bibermann IS Farooqi et al, MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med 24: 551-555.
  63. Sarkar S, Legradi G, Lechan RM. 2002 Intracerebroventricular administration of -melanocyte stimulating hormone increases phosphorylation of CREB in TRH-producing and CRH-producing neurons of the hypothalamic pavaventricular nucleus. Brain Res 945: 50-59.
  64. Hahn TM, Breininger JF, Baskin DG, Schwartz MW. 1998 Coexpression of Agrp and NPY in fasting-activated hypothalalmic neurons. Nat Neurosci 1: 271-272.
  65. Spiegelman BM, Flier JS. 2001 Obesity and the regulation of energy balance. Cell 104: 531-543.
  66. Wirth MM, Olszewski PK, Yu C, Levine AS, Giraudo SQ. 2001 Paraventricular hypothalamic alpha-melanocyte-stimulating hormone and MTII reduce feeding without causing aversive effects. Peptides 22: 129-134.
  67. Giraudo SQ, Billington CJ, Levine AS. 1998 Feeding effects of hypothalamic injection of melanocortin 4 receptor ligands. Brain Res 809: 302-306.
  68. Luiten PG, ter Horst GJ, Karst H, Steffens AB. 1985 The course of paraventricular hypothalamic efferents to autonomic structures in medulla and spinal cord. Brain Res 329: 374-378.
  69. Roeling TA, Veening JG, Peters JP, Vermelis ME, Nieuwenhuys R. 1993 Efferent connections of the hypothalamic “grooming area” in the rat. Neuroscience 56: 199-225.
  70. Sawchenko PE, Swanson LW. Immunohistochemical identification of neurons in the paraventricular nucleus of the hypothalamus that project to the medulla or to the spinal cord in the rat. J Comp Neurol 1982; 205:260-272.
  71. Haynes WG, Morgan DA, Djalali A, Sivitz WI, Mark AL. 1999 Interactions between the melanocortin system and leptinin control of sympathetic nerve traffic. Hypertension 33: 542-547.
  72. Bamshad M, Aoki VT, Adkison MG, Warren WS, Bartness TJ. 1998 Central nervous system origins of the sympathetic nervous system outflow to white adipose tissue. Am J Physiol 275: R291-R299.
  73. Shah BP, Vong L, Olson DP, Koda S, Krashes MJ, Ye C, Yang Z, Fuller PM, Elmquist JK, Lowell BB.2014 MC4R-expressing glutamatergic neurons in the paraventricular hypothalamus regulate feeding and are synaptically connected to the parabrachial nucleus.  Proc Natl Acad Sci USA 111: 13193-13198.
  74. Carter ME, Soden ME, Zweifel LS, Palmiter RD. 201. Genetic identification of a neural circuit that suppresses appetite.Nature 503: 111-114.
  75. Blouet C, Jo YH, Li X, Schwaratz GJ. 2009 Mediobasal hypothalamic leucine sensing regulates food intake through activation of a hypothalamus-brainstem circuit. J Neurosci 29: 8302-8311.
  76. Singru PS, Wittmann G, Farkas E, Zseli G, Fekete C, Lechan RM. 2012 Refeeding-activated glutamatergic neurons in the hypothalamic paraventricular nucleus (PVN) mediate effects of melanocortin signaling in the nucleus tractus solitarius (NTS). Endocrinology 153: 3804-3814.
  77. Blevins JE, Morton GJ, Williams DL, Caldwell DW, Bastian LS, Wisse BE, Schwartz MW, Baskin DG. 2009 Forebrain melanocortin signaling enhances the hindbrain satiety reponse to CCK-8. Am J Physiol Regul Integr Comp Physiol 296: R476-484.
  78. Blevins JE, Schwartz MW, Baskin DG. 2004 Evidence that paraventricular nucleus oxytocin neurons link hypothalamic leptin action to caudal brain stem nuclei controlling meal size. Am J Physiol Regul Integr Comp Physiol 287: R97-96.
  79. Elmquist JK. 2001 Hypothalamic pathways underlying the endocrine, autonomic, and behavioral effects of leptin. Phys Behav 74: 703-708.
  80. Saper CB. 1985 Organization of cerebral cortical afferent systems in the rat. II. Hypothalamocortical projections. J Comp Neurol 237: 21-46.
  81. Qu D, Ludwig DS, Gammeltoft S, Piper M, Pelleymounter MA, Cullen MJ, Mathes WF, Przypek R, Kanarek R, Maratos-Flier E. 1996 A role for melanin-concentrating hormone in the central regulation of feeding behaviour, Nature 380: 243-247.
  82. Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E. 1998 Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature 396: 670-674.
  83. Sakurai T. 2002 Roles of orexins inregulation of feeding and wakefulness. Neuroreport 13: 987-995.
  84. Krowicki ZK, Burmeister MA, Berthoud HR, Scullion RT, Fuchs K, Hornby PJ. 2002 Orexins in rat dorsal motor nucleus of the vagus potently stimulate gastric motor function. Am J Physiol Gasstrointest Liver Physiol 283: G465-472.
  85. Elias CF, Lee C, Kelly J, Aschkenasi C, Ahima RS, Couceyro PR, Kuhar MJ, Saper CB, Elmquist JK. 1998 Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron 21: 1375-1385.
  86. Ahima RS, Flier JS. 2000 Leptin. Annu Rev Physiol 62: 413-437.

399 Morton GJ. Niswender KD, Rhodes CJ, Myers MG, Blevins JE, Baskin DG, Schwartz MW. 2003 Arcuate nucleus-specific leptin receptor gene therapy attenuates the obesity phenotype of Koletsky (fa(k)/fa(k)) rats. Endocrinology 144: 2016-2024.

  1. Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, Tang V, Liu SM, Ludwig T, Chua SC Jr, Lowell BB, Wlmquist JK. 2005 The hypothalamic arcuate nucleus: a key site for mediating leptin’s effects on glucose homeostasis and locomotor activity. Cell Metab 1: 63-72.
  2. ter Horst GJ, Luiten PGM. 1986 The projections of the dorsomedial hypothalamic nucleus in the rat. Brain Res Bull 16: 231-248.
  3. Bellinger LL, Bernardis LL. 2002 The dorsomedial hypothalamic nucleus and its role in ingestive behavior and body weight regulation: lesions learned from lesioning studies. Physiol Behav 76: 431-442.
  4. Bellinger LL, Bernardis LL, Williams FE. 1984 Suppressionof feeding b y cholecystokinin but not bombesin is attenuated by dorsomedial hypothalamic nuclei lesioned rats. Peptides 5: 547-552.
  5. Fulwiler CE, Saper CB. 1985 Cholecystokinin-immunoreactive innervation of the ventromedial hypothalamus in the rat: possible substrate for autonomic regulation of feeding. Neurosci Lett 53: 289-296.
  6. Choi S, Wong LS, Yamat C, Dallman MF. 1998 Hypothalamic ventromedial nuclei amplify circadian rhythms: Do they contain a food-entrained endogenous oscillator? J Neurosci 18: 3843-3852.
  7. Grill HJ, Schwartz MW, Kaplan JM, Foxhall JS, Breininger J, Baskin DG. 2002 Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology 143: 239-246.
  8. Goldstone AP, Mercer JG, Gunn I, Moar KM, Edwards CM, Tossi M, Howard JK, Rasheed S, Turton MD, Small C, Heath MM, O’Shea D, Steere J, Meeran K, Ghatei MA, Hoggard N, Bloom SR. 1997 Leptin interacts with glucagons-like peptide-1 neurons to reduce food intake and body weight in rodents. FEBS Lett 415: 134-138.
  9. Al-Barazanji KA, Arch JR, Buckingham RE, Tadayyon M. 2000 Central exendin-4 infusion reduces body weight without altering plasma leptin in (fa/fa) Zucker rats. Obes Res 8: 317-323.
  10. Merchenthaler I, Lane M, Shughrue P. 1999 Distribution of pre-pro-glucagon and glucagons-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol 403: 261-280.
  11. Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ, Taylor GM, Heath MM, Lambert PD, Wilding JP, Smith DM, Ghatei MA, Herbert J, Bloom SSR. 1996 A role for glucagons-like peptide-1 in the central regulation of feeding. Nature 379: 69-72.
  12. Larsen PJ, Tang-Christensen M, Holst JJ, Orskov C. 1997 Distribution of glucagons-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience 77: 257-270.
  13. McMahon LR, Welllman PJ. 1998 PVN infusion of GLP-1-(7-36) amide suppresses feeding but does not induce aversion or alter locomotion in rats. Am J Physiol 274: R23-R29.
  14. Hayes MR, Skibicka KP, Leichner TM, Guarnieri DJ, DiLeone RJ, Bence KK, Grill HJ. 2010 Endogenous leptin signaling in the caudal nucleus tractus solitarius and area postrema is required for energy balance regulation. Cell Metab 11: 77-83.
  15. Hommel JD, Trinko R, Sears RM, Georgescu D, Liu ZW, Gao XB, Thurmon JJ, Marinelli M, DiLeone RJ. 2006 Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron 51: 801-810.
  16. Naslund E, Hellstrom PM. 2007 Appetite signaling: From gut peptides and enteric nerves to brain. Physiol. Behav. 92: 265-262.
  17. Suzuki K, Jayasena CN, Bloom SR. 2012 Obesity and appetite control. Exp Diabetes Res, Article ID 824305, 1-19.
  18. Bauer PV, Hamr SC, Duca FA. 2016. Regulation of energy balance by a gut-brain axis and involvement of the gut microbiota.Cell Mol Life Sci 73: 737-755.
  19. Murphy KG, Dhillo WS, Bloom SR. 2006 Gut peptides in the regulation of food intake and energy homeostasis. Endocrine Reviews 27: 719-727.
  20. Schwartz MW, Sipols AJ, Marks JL, Sanacora G, White JD, Scheurink A, Kahn SE, Baskin DG, Woods SC, Figlewicz DP. 1992 Inhibition of hypothalamic neuropeptide Y gene expression by insulin. Endocrinology 130: 3608-3616.
  21. Woods SC, Seeley RJ, Porte D Jr, Schwartz MW. 1998 Signals that regulate food intake and energy homeostasis. Science 280: 1378-1383.
  22. Reda TK, Geliebter A, Pi-Sunyer FX. 2002 Amylin, food intake, and obesity. Obes Res 10: 1087-1091.
  23. Schwartz GJ, McHugh PR, Moran TH. 1993 Gastric loads and cholecysstokinin synergistically stimulate rat gastric vagal afferents. Am J Physiol 265: R872-R876.
  24. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE, Low MJ, Ghatel M, Cone RD, Bloom SR. 2002 Gut hormone PYY3-36 physiologically inhibits food intake. Nature 418: 650-654.
  25. Abbot CR, Monteiro M, Small CJ, SajediA, Smith KL, Parkinson JR, Ghatei MA, Bloom SR. 2005 The inhibitory effects of peripheral administration of peptide YY (3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 1044:127-131.
  26. Baggio LL, Huang Q, Brown TJ, Drucker DJ. 2004 Oxntomodulin and glucagon-like peptide-1 differently regulate murine foodintake and energy expenditure. Gastroenterology 127: 546-558.
  27. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, Bloom SR. 2006 Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomized controlled trial.Int. J. Obes.  30: 1729-1736.
  28. Asakawa A, Inui A, Yuzuriha H, Ueno N, Katsuura G, Fujimiya M, Fujino MA, Niijima A, Meguid MM, Kasuga M. 2003. Characterization of the effects of pancreatic plypeptide in the regulation of energy balance. Gastroenterology 124: 1325-1336.
  29. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. 2001 A preprandial rise inplasma ghrelin levels suggests a arole in meal initiation in humans. Diabetes 50: 1714-1719.
  30. Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, Wakabayashi I. 2001 Chronic central infusion of ghrelin increases hypothalamic neuropeptide Y and agouti-related protein mRNA levels and body weight in rats. Diabetes 50: 2438-2443.
  31. Williams KW, Elmquist JK. 2012 From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat Neurosci 15: 1530-1355.
  32. Williams DL, Grill HJ, Cummings DE, Kaplan JM. 2003 Vagotomy dissociates short- and long-term controls of circulating ghrelin. Endocrinology 144: 5184-5187.
  33. Cummings DE, Clement K, Purnell JQ, Vaisse C, Foster KE, Frayo RS, Schwartz MW, Basdevant A, Weigle DS. 2002 Elevated plasma ghrelin levels in Prader Willi syndrome. Nat Med. 8:643-4.
  34. Goldstone AP, Thomas EL, Brynes AE, Castroman G, Edwards R, Ghatei MA, Frost G, Holland AJ, Grossman AB, Dorbonits M, Bloom SR, Bell JD. 2004 Elevated fasting plasma ghrelin in Prader-Willi syndrome adults is not solely explained by their reduced visceral adiposity and insulin resistance. J Clin. Endocrinol. Metab. 89: 1718-1726.
  35. Zhang JV, Ren PG, Avsian-Kretchmer O, Luo GW, Rauch R, Klein C, Hsueh AJ. 2005 Obestatin, a peptide encoded by the ghrelin gene, oposes ghrelin’s effects on food intake. Science 310: 996-999.
  36. Stengel A, Gobel M, Yakubov I, Wang L, Witcher D, Coskun T, Tache Y, Sachs G, Lambrecht NW. 2008 Identification and characterization of nesfatin-1 immunoreactivity in endocrine cell types of the rat gastric oxyntic mucosa. Endocrinology, in press.
  37. Oh-I S, Shimizu H, Satoh T, Okada S, Adachi S, Inoue K, Eguchi H, Yamamoto M, Imaki T, Hashimoto K, Tsuchiya T, Monden T, Horiguchi K, Yamada M, Mori M. 2006 Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature 443: 709-712.
  38. Price CJ, Samson WK, Ferguson AV. 2008 Nesfatin-1 inhibits NPY neurons in the arcuate nucleus. Brain Res 1230: 99-106.
  39. Fort P, Salvert D, Hanriot L, Jego S, Shimizu H, Hashimoto K, Mori M, Luppi PH. 2008 Neuroscience The satiety molecule nesfatin-1 is co-expressed with melanin concentrating hormone in tuberal hyhpothalamic neurons of the rat. Neuroscience 155: 174-181.
  40. Gavrieli A, Mantzoros CS. 2016. Novel molecules regulating energy homeostasis: physiology and regulation by macronutrient intake and weight loss. Endocrinology and Metabolism 31: 361-372.
  41. Abraham G, Falcou R, Rozen R, Mandenoff A, Autissier N, Apfelbaum M. 1987 The effects of a constant T3 level and thermoneutrality in diet-induced hyperphagia. Horm Metab Res 19(3): 96-100.
  42. Syed MA, Thompson MP, Pachucki J, Burmeister LA. 1999 The effect of thyroid hormone on size of fat depots accounts for most of the changes in leptin mRNA and serum levels in the rat. Thyroid 9(5): 503-512.
  43. Ishii S. Kamegai J, Tamura H, Shimizu T, Sugihara H, Oikawa S. 2003 Hypothalamic neuropeptide Y/Y1 receptor pathway activated by a reduction in circulating leptin, but not by an increase in circulating ghrelin, contributes to hyperphagia associated with triiodothyronine-induced thyrotoxicosis. Neuroendocrinology 78(6): 321-230.
  44. Luo L, MacLean DB. 2003 Effects of thyroid hormone on food intake, hypothalamic Na/K ATPase activity and ATP content. Brain Res 973(2): 233-239.

444  Kong W M, Martin NM, Smith KL, Gardiner JV, Connoley IP, Stephens DA, Dhillo WS, Ghatei MA, Small CJ, Bloom SR. 2004 Tri-iodothyronine Stimulates Food Intake via the Hypothalamic Ventromedial Nucleus Independent of Changes in Energy Expenditure. Endocrinology 145: 5252-5258.

  1. Coppla A, Liu ZW, Andrews ZB, Paradis E, Roy MC, Friedman JM, Ricquier D, Richard D, Horvath TL, Gao XB, Diano S. 2007 A central thermogenic-like mechanism in feeding regulation: an interplay between arcuate nucleus T3 and UCP2.Cell Metab. 5: 21-33.
  2. Puymirat, J. 1992 Thyroid receptors in the rat brain. Prog Neurobiol 39(3): 281-294.
  3. Lechan RM, Qi Y, Jackson IMD, Mahdavi V. 1994 Identification of thyroid hormone receptor isoforms in thyrotropin-releasing hormone neurons of the hypothalamic paraventricular nucleus. Endocrinology 135(1): 92-100.
  4. Broedel O, Eravci M, Stoltenburg-Didinger G, Plueckhan H, Fuxius S, Meinhold H, Baumgartner A. 2003 Effects of hyper- and hypothyroidism on thyroid hormone concentrations in regions of the rat brain. Am J Physiol Endocrinol Metab 285(3): E470-E480.
  5. Campos-Barros A, Musa A, Flechner A, Hessenius C, Gaio U, Meinhold H, Baumgartner A. 1997 Evidence for circadian variations of thyroid hormone concentrations and type II 5'-iodothyronine deiodinase activity in the rat central nervous system. J Neurochem 68(2): 795-803.
  6. Heine A, Taylor JA, Iwamoto GS, et al. 2000. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA, 97: 12729-12734.
  7. Xu Y, Nedungadi TP, Zhu L, et al. 2011. Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metab 14: 453-465.
  8. Kadowaki T, Yamauchi T, Kubota N. 2008 The physiological and pathophysiological role of adiponectin and adiponectin receptors in the peripheral tissues and CNS. FEBS Lett 582: 74-80.

453 Kubota N, Yano W, Kubota T, Yamauchi T, Itoh S, Kumagai H, Kozono H, Takamoto I, Okamoto S, Shiuchi T, Suzuki R, Satoh H, Tsuchida A, Moroi M, Sugi K, Noda T, Ebinuma H, Ueta Y, Kondo T, Araki E, Ezaki O, Nagai R, Tobe K, Terauchi Y, Ueki K, Minokoshi Y, Kadowaki T. 2007 Adiponectin stimulates AMP-activated protein kinase in the hypothalamus and increases food intake. Cell Metab 6: 55-68.

  1. Lechan RM, Fekete C. 2010 Neuroendocrine and metabolic adaptations in the central nervous system to weight loss that facilitate weight re-gain. Pediatric Obesity: Etiology, Pathogenesis, and Treatment, M. Freemark, Ed, Humana Press/Springer, pp
  2. Williams KW, Elmquist JK. 2012 From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat Neurosci 15: 1350-1355.
  3. Piomeilli D. 2003 The molecular logic of endocannabinoids signaling. Nat Rev Neurosci 4: 873-884.
  4. Wittmann G, Deli L, Kallo I, Hrabovszky E, Watanabe M, Liposits Z, Fekete C. 2007 Distribution of type 1 cannabinoid receptor (CB1)-immunoreactive axons in the mouse hypothalamus. J Comp Neurol 10: 270-279.
  5. Freund TF, Katona I, Piomelli D. 2003 Role of endogenous cannabinoids in synaptic signaling. Physiol Rev 83: 1017-1066.
  6. Kola B, Farkas I, Christ-Crain M, Wittmann G, Lolli F, Amin F, Harvey-White J, Liposits Z, Kunos G, Frossman AB, Fekete C, Korbonits M. 2008 The orexigenic effect of ghrelin is mediated through central activationof the endogenous cannabinoid system. PLoS One 3:e1797.
  7. Jo YH, Chen YJ, Chua SC Jr, Talmage DA, Role LW. 2005 Integration of endocannabinoids and leptin signaling in an appetite-related neural circuit. Neuron 48: 1055-1066.
  8. Tong Q, Ye CP, Jones JE, Elmquist JK, Lowell BB. 2008 Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat Neurosci 11: 998-1000.
  9. Vong L, Ye C, Yang Z, Choi B, Chua S Jr, Lowell BB. 2011 Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 14: 142-154.
  10. Xu Y, Berglund ED, Sohn JW, Holland WL, Chuang JC, Fukuda M, Rossi J, Williams KW, Jones JE, Zigman JM, Lowell BB, Sherer PE, Elmquist JK. 2010 5-HT2CRs expressed by pro-opiomelanocortin neurons regulate insulin sensitivity in liver. Nat Neurosci 13: 1457-1459.
  11. Xu Y, Jonwa JE, Kohno S WR. 2008 L 5-HT2CRs expressed by pro-opiomenalocortin neurons regulate energy homeostasis. Neuron 60: 582-589.
  12. Burke LK, Heisler LK. 2015. 5-hydroxytryptamine medications sfor the treatment of obesity. J Neuroendocrinol 27: 389-398.
  13. Jarvie BC, Hentges ST. 2012 Expression of GABAergic and glutamatergic phenotypic markers in hypothalamic proopiomelanocortin neurons. J Comp Neurol 520: 3863-3876.
  14. Fulton S, Pissios P, Manchon RP, Stiles L, Frank L, Pothos EN, Maratos-Flier E, Flier JS. 2006 Leptin regulation of the mesoaccumbens dopamine pathway. Neuron 51: 811-822.
  15. Kelly AE, Berridge KC. 2002 The neuroscience of natural rewards: relevance to addictive drugs. J Neurosci 22: 3306-3311.
  16. Geiger BM, Haburcak M, Avena NM, Moyer MC, Hoebel BG, Pothos EN. 2009 Deficits of mesolimbic dopamine neurotransmission in rat dietary obesity. Neuroscience 159: 1193-1199.
  17. Ritter S, Dinh TT, Li AJ. 2006 Hindbrain catecholamine neurons control multiple glucoregulatory reponses. Physiol. Behav. 89: 490-500.
  18. Cota D, Proulx K, Smith KA, Kozma SC, Thomas G, Woods SC, Seeley RJ. 2006 Hypothalamic mTOR signaling regulates food intake. Science 312: 927-930.
  19. Blouet C, Schwartz GJ. 2012 Brainstem nutrient sensing in the nucleus of the solitary tract inhibits feeding. Cell Metab 16: 579-587.
  20. Kim EK, Miller I, Aja S, Landree LE, Pinn M, McFadden J, Kuhajda FP, Moran TH, Ronnett GV. 2004 C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase. J. Biol. Chem. 279: 19970-19976.
  21. Ibrahim N, Bosch MA, Smart JL, Qui J, Rubinstein M, Ronnekleiv OK, Low MJ, Kely MJ. 2003 Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. Endocrinology 144:1331-1340.
  22. Parton LE, Ye CP, Coppari R, Enriori PJ, Choi B, Zhang C-Y, Xu C, Vianna CR, Balthasar N, Lee CE, Elmquist JK, Clwley MA, Lowell BB. 2007 Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature 449: 228-232.
  23. Farr OM, Li CR, Mantzzoros CS. 2016. Central nervous system regulation of eating: insights from human brain imaging. Metabolism 65: 699-713.
  24. Freeman ME, Kanyicska B, Lerant A, Nagy G. 2000 Prolactin: Structure, function, and regulation of secretion. Phys Rev 80: 1523-1631.
  25. Dubois-Dauphin M, Armstrong W, Tribollet E, Drefuss JJ. 1985 Somatosensory systems and the milk-ejection reflex in the rat. I. Lesions of the mesencephalic lateral tegmentum disrupt the reflex and damage mesencephalic somatosensory connections. Neuroscience 15: 1111-1129.
  26. Dubois-Dauphin M, Armstrong W, Tribollet E, Drefuss JJ. 1985 Somatosensory systems and dthe milk-ejection reflex in the rat. II. The effects of lesions in the ventroposterior thalamic complex, dorsal columns and lateral cervical nucleus-dorsolateral funiculus. Neuroscience 15: 1131-1140.
  27. Crowley WR. 2015 Neuroendocrine regulation of lactationand milk production. Compr Physiol 5:255-291.
  28. Li C, Chen P, Smith MS. 1999 Neural populations in the rat forebrain and brainstem activated by the suckling stimulus as demonstrated by cFos expression. Neuroscience 94: 117-129.
  29. Grattan DR. 2002 Behavioral significance of prolactin signaling in the central nervous sysem during pregnancy and lactation. Reproduction 123: 497-506.
  30. Ben-Jonathan N, Hnasko R. 2001 Dopamine as a prolactin (PRL) inhibitor. Endocr Rev 22: 724-763.
  31. Romano Y, Yip SH, Hodson DJ, Guillou A, Parnaudeau S, Kirk S, Tronche F, Bonnefort X, Le Tissier P, Bunn SJ, Grattan DR, Mollard P, Martin AO. Plasticity of dopamine neurons during lactation results in dissociation of electrical activity and release. J Neurosci 33: 4424-4433.
  32. Walsh RJ, Slaby FJ, Posner BI. 1987 A receptor-mediated mechanism for the transport of prolactin from blood to cerebrospinal fluid. Endocrinology 120: 1846-1850.
  33. Pi XJ, Grattan DR. 1999 Increased expression of both short and long forms of prolactin receptor RNA in hypothalamic nuclei of lactating rats. J Neuroend 11: 693-705.
  34. Bridges RS, Rigero BA, Byrnes EM, Yang L, Walker AM. 2001 Central infusions of the recombinant human prolactin receptor antagonist, S179D-PRL, delay the onset of maternal behavior in steroid-primed, nulliparous female rats. Endocrinology 142: 730-739.
  35. Yamada S., Uenoyama Y, Kinoshita M, Iwata K, Takase K, Matsui H, Adachi S, Inoue K, Maeda K-I, Tsukamura H. 2007 Inhibition of metastin (kisspeptin-54)-GPR54 signaling in the arcuate nucleus-median eminence region during lactation in rats, Endocrinology 148: 2226-2232.
  36. Smith MS, True C, Grove KL. 2010 The neuroendocrine basis of lactation-induced suppressionof GnRH: role of kisspeptin and leptin. Brain Res 1364: 139-152.
  37. Li C, Chen P, Smith MS. 1998 The acute suckling stimulus induces expression of neuropeptide Y (NPY) in cells in the dorsomedial hypothalamus and increases NPY expression in the arcuate nucleus. Endocrinology 139: 1645-1652.
  38. Smith MS. 1993 Lactation alters neuropeptide-Y and proopiomelanocortin gene expression in the arcuate nucleus of the rat. Endocrinology 133: 1258-1265.
  39. Smith MS, Grove KL. 2002 Integration of the regulation of reproductive function and energy balance: lactation as a model. Front Neuroend 23: 225-256.
  40. Chen P, Li C, Haskell-Luevano C, Cone R, Smith MS. 1999 Altered expression of agouti-related protein (AGRP) and its colocalization with neuropeptide Y (NPY) in the arcuate nucleus of the hypothalamus during lactation. Endocrinology 140: 2645-2650.
  41. Li C, Chen PL, Smith MS. 1999 Identification of neuronal input to the arcuate nucleus (ARH) activated during lactation: implications in the activation of neuropeptide Y neurons. Brain Res 824: 267-276.
  42. Li C, Chen P, Smith MS. 1999 Morphological evidence for direct interaction between arcuate nucleus neuropeptide Y (NPY) neurons and gonadotropin releasing hormone (GnRH) neurons and the possible involvement of Npy Y1 receptors. Endocrinology 140: 5382-5390
  43. Campbell RE, French-Mullen JMH, Cowley MA, Smith MS, Grove KL. 2001 Hypothalamic circuitry of neuropeptide Y regulation of neuroendocrine function and food intake via the Y5 receptor sybtype. Neuroendocrinology 74: 106-119.
  44. Theodosis DT, Poulain DA, Olier SHR. 2008. Activity-dependent structural and functional plasticity of astrocyte-neuron interactions. Physiol Rev 88: 983-1008.
  45. Nakamura K. 2011 Central circuitries for body temperature regulation and fever. Am J Physiol Regul Integr Comp Physiol 301: R1207-1228.
  46. Morrison SF. May 2016 Central control of body temperature. F1000Research, Published online
  47. Siemens J, Kamm GB. 2018. Cellular populations and thermosensing mechanisms of the hypothalamic thermoregulatory center. Pflugers Arch 470:809-822.
  48. Boulant JA. 2000 Role of the preoptic-anterior hypothalamus in thermoregulation and fever, Clin Inf Dis 31: S157-167.
  49. Chen X-M, Hosono T, Yoda T, Fukuda Y, Kanosue K. 1998 Efferent projection from the preoptic area for the control of nonshivering thermogenesis in rats. J Physiol 512: 883-892.
  50. Hefco E, Krulich L, Illner P, Larsen PR. 1975 Effect of acute exposure to cold on the activity of the hypothalamic-pituitary-thyroid system, Endocrinology 97: 1185-1195.
  51. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, Kolodny GM, Kahn CR. 2009 Identification and importance of brown adipose tissue in adult humans. N Engl J Med 360: 1509-1517.
  52. Nedergaard J, Bengtsson T, Cannon B. 2007 Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 293: E444-452.
  53. Cypess AM, Kahn CR. 2010 The role and importance of brown adipose tissue in energy homeostasis. Curr Opin Pediatr 22: 478-484.
  54. Tam CS, Lecoultre V, Ravussin E. 2012 Brown adipose tissue: mechanisms and potential therapeutic targets. Circulation 125: 2782-2791.
  55. Kanosue K, Ahang Y-H, Yanase-Fujiwara M, Hosono T. 1994 Hypothalamic network for thermoregulatory shivering, Am J Physiol 267: R275-R282.
  56. Nagashima K, Nakai S, Tanaka M, Kanosue K. 2000 Neuronal circuitries involved in thermoregulation, Auton Neurosci 85: 18-15.
  57. Bamshad M, Song CK, Bartness TJ. 1999 CNS origins of the sympathetic nervous system outflow to brown adipose tissue. Am. J. Physiol. 276: R1568-R1578.
  58. Yoshida K, Maruyama M, Hosono T, Nagashima K, Fukuda Y, Gerstberger R, Kanosue K. 2002 Fos expression induced by warming the preoptic area in rats, Brain Res 933: 109-117.
  59. Zhang Y-H, Hosono T, Yanase-Fujiwara M, Chen X-M, Kanosue K. 1997 Effect of midbrain stimulation on thermoregulatory vasomotor responses in rats, J Physiol 503: 177-186.

513 Tanaka M, Nagashima K, McAllen RM, Kanosue K. 2002 Role of the medullary raphe in thermoregulatory vasomotor control in rats, J Physiol 540: 657-664.

  1. Rathner JA, Owens NC, McAllen RM. 2001 Cold-activated raphe-spinal neurons inrats, J Physiol 535: 841-854.
  2. Nakamura K. 2011 Central circuitries for body temperature regulation and fever. Am J Physiol Regul Integr Comp Physiol 301:R1207-1228.
  3. Caterina MJ. 2007 Transient receptor potential ion channels as participants in thermosensation and thermoregulation. Am. J. Physiol. Regulatory Integrative Comp. Physiol. 292: 64-76.
  4. Jancso-Gabor A, Szolcsanyi J, Jancso N. 1970 Stimulation and desensitization of the hypothalamic heat-sensitive sructures by capsaicin in rats. J. Physiol 208: 449-459.
  5. Sasamura T., Sasaki M., Tohda C., Kuraishi Y. 1998 Existence of capsaicin-sensitive glutamatergic tgerminals in rat hypothalamus. Neuroreport 9: 2045-2048.
  6. Szelenyi Z, Hummel Z, Szolcsanyi J, Davis JB. 2004 Daily body temperature rhythm and heat tolerance in TRPV1 knockout and capsaicin-pretreated mice. Eur. J. Neurosci 19: 1421-1424.
  7. Song K, Wang H, Kamm GB et al. 2016. The TRPM2 channel is a hypothalamic heat sensor that limits fever and can drive hypothermia Science 353: 1393-1398.
  8. Lu J, Zhang Y-H, Chou TC, Gaus SE, Elmquist JK, Shiromani P, Saper CB. 2001 Contrasting effects of ibotenate lesions of the paraventricular nucleus and subparaventricular zone on sleep-wake cycle and temperature regulation, J Neurosci 21: 4864-4874.
  9. Watts AG. The efferent projections of the suprachiasmatic nucleus: anatomical insights into the control of circadian rhythms. In: Suprachiasmatic nucleus: the mind’s clock, Klein DC, Moore RY, Reppert SM, Eds, New York, Oxford UP, pp 77-106, 1991.
  10. Kolka M, Stephenson L. 1989 Control of sweating during the human menstrual cycle, Eur J Appl Physiol 58: 890-895.
  11. Stephenson LA, Kolka MA. 1999 Esophageal temperature threshold for sweating decreases before ovulation in premenopausal women, J Appl Physiol 86: 22-28.
  12. Silva NL, Boulant JA. 1986 Effects of testosterone, estradiol, and temperature on neurons in preoptic tissue slices, Am J Physiol 250: R625-R632.
  13. Nakayama T, Suzuki M, Ishizuka N. 1975 Action of progesterone on thermosensitive neurons, Nature 258: 80.
  14. McEwen BS, 1981 Neural gonadal steroid actions, Science 211: 1303-1311.
  15. Freedman RR. 2001 Physiology of hot flashes, Am J Human Biol 13: 453-464.
  16. Freedman RR. Woodward S. 1995 Altered shivering threshold in postmenopausal women with hot flashes, Menopause 2: 163-168.
  17. Freedman RR, Dinsay R. 2000 Clonidine raises the sweating threshold in symptomatic but not in asymptomatic postmenopausal women. Fertil Steril. 74:20-23.
  18. Dacks PA, Krajewski SJ, Rance NE. 2011. Activation of neurokinin 3 receptors in the median preoptic nucleus decreases core temperature in the rat. Endocrinology 152:4894-4905.
  19. Jayasena CN, Comminos AN, De Silva A, et al, 2014 Effects of neurokinin B administration on reproductive hormone secretion in healthy men and women. J Clin Endocrinol Metab 99: E19-E27.
  20. Prague JK, Roverts RE, Comminos AN, et al. 2018 Neurokinin 3 receptor antagonism rapidly improves vasomotor symptoms with sustained duration of action. Menopause 25:862-869.
  21. Skorupskaite K, George JT, Velldhuis JD et al. 2018 Neurokinin 3 receptor antagonism reveals roles for neurokinin B in the regulation of gonadotropin secretion and hot flashes in postmenopausal women. Neuroendocrinology 106: 148-157.
  22. van det Heide A, Werth E, Donjacour CE et al. 2016 Core body and skin temperature in type 1 narcolepsy in daily life; effects of sodium oxybate and prediction of sleep attacks. Sleep 2016 39: 1941-1949.
  23. Kluger MJ. The adaptive value of fever. In: Mackowiak PA, Ed, Fever: basic mechanisms and management, New York, Raven Press, pp 105-124, 1991.
  24. Duff GW. 1986 Is fever beneficial to the host: a clinical perspective, Yale J Biol Med 59: 125-130.
  25. Scammell TE, Saper CB. 1997 Mechanisms of CNS response to systemic immune challenge: the febrile response, Trends Neurosci 20: 565-570.
  26. Saper CB, NeurologicalTE, Jacobson CD, Saper CB. 1996 Distribution of fos-like immunoreactivity in the rat brain following intravenous lipopolysaccharide administration, J Comp Neurol 371: 85-103.
  27. Lazarus M, Yoshida K, Coppari R, Bass CE, Mochizuki T, Lowell BB, Saper CB. 2007 EP3 prostaglandin receptors in the median preoptic nucleus are critical for fever responses. Nat. Neurosci. 10: 1131-1133.
  28. Nakamura Y, Nakamura K, Matsumura K, Kobayashi S, Keneko T, Morrison SF. 2005 Direct pyrgenic input fromprostaglandin EP3 receptor-expressing preoptic neurons to the dorsomedial hypothalamus. Eur. J. Neurosci. 22: 3137-3146.
  29. Morrison SF, Nakamura K, Madden CJ. 2008 Central control of thermogenesis in mammals. Exp. Physiol. 93: 773-797.
  30. Blatteis CM, Sehic E, Li S. 1998 Afferent pathways of pyrogen signaling, Ann NY Acad Sci 856:95-107.
  31. Ek M, Kurosawa M, Lundeberg T, Ericsson A. 1998 Activation of vagal afferents after intravenous injection of interleukin-1b: role of endogenous prostaglandins, J Neurosci 18: 9471-9479.
  32. Huang Q-H, Hruby VJ, Tatro JB. 1998 Systemic -MSH suppresses LPS fever via central melanocortin receptors independently of its suppression of corticosterone and IL-6 release, Am J Physiol 275: R524-R530.
  33. Huang Q-H, Entwistle ML, Alvaro JD, Duman RS, Hruby VJ, Tatro JB. 1997 Antipyretic role of endogenous melanocortins mediated by central melanocortin receptors during endotoxin-induced fever, J Neurosci 17: 3343-3351.
  34. Jacobowitz DM, O’Donohue TL. 1978 -Melanocyte stimulating hormone: immunohistochemical identification and mapping in neurons of rat brain, Proc Natl Acad Sci 75: 6300-6304.
  35. Moore R. Y. 1999 Circadian timing, in Zigmond M.J., Bloom F.E., Landis S. C., Robetos J. L., Squire L. R. (eds), Fundamental Neuroscience, Academic Press, pp. 1189-1206.
  36. Kriegsfeld LJ, LeSaute J, Hamada T, Pitts SM, Silver R. 2002 Circadian rhythms in the endocrine system, in Pfaff D. W, Arnold A.P., Etgen A.M., Fahrback S.E., Rubin R.T. (eds), Hormones, Brain and Behavior, vol 2, Academic Press, pp.33-91.
  37. Welch DK, Lagothetis DE, Meister M, Reppert SM. 1995 Individual neurons dissociated from rat suprachiasmatic nucleus express independently phased circadian firing patterns. Neuron 14: 697-706.
  38. Shirakawa T, Honma S, Honma K. 2001 Multiple oscillators in the suprachiasmatic nucleus. Chronobiol Int 18: 371-387.
  39. Moore R.Y. 1983 Organization and function of a central nervous systems circadian oscillator: the suprachiasmatic hypothalamic nucleus. Fed. Proc 43: 2783-2789.
  40. Ibuka M, Inoue S-L, Kawamura H. 1977 Analysis of sleep-wakefulness rhythms in male rats after suprachiasmatic lesions and ocular enucleation. Brain Res. 122: 33-47.
  41. Ralph MR, Foster RG, Davis FC, Menaker M. 1990 Transplanted suprachiasmatic nucleus determines circadian rhythms. Science 247: 975-978.
  42. Mehta N, Cheng HY. 2012 Micro-Managing the Circadian Clock: The Role of microRNAs in Biological Timekeeping. J Mol Biol. 2012 Nov 6. doi:pii: S0022-2836(12)00855-8. 10.1016/j.jmb.2012.10.022. [Epub ahead of print]
  43. van Ooijen G, Millar AJ. 2012. Non-transcriptional oscillators in circadian timekeeping Trends Biochem Sci. 37:484-492.
  44. Mrosovsky N.1996 Locomotor activity and non-photic influences on circadian clocks.  Biol Rev Camb Philos Soc 71: 343-372.
  45. Zee PC, Manthena P.2007 The brain’s master circadian clock:  implications and opportunities for therapy of sleep disorders.  Sleep Med Rev 11: 59-70.
  46. Tonsfeldt KJ., Chappell PE.2012Clocks on top:  the role of the circadian clock in the hypothalamic and pituitary regulation of endocrine physiology. Mol Cell Endocrinol   349: 3-12.
  47. Cheng, M.Y., Bullock, C.M., Li, C., Lee, A.G., Bermak, J.C., Belluzzi, J., Weaver, D.R., Leslie, F.M., Zhou, Q.Y. 2002. Prokineticin 2 transmits the behavioural circadian rhythm of the suprachiasmatic nucleus. Nature 417, 405–410.
  48. Hamada T, LeSauter J, Venuti JM, Silaver R. 2001 Espression of Period genes: rhythmic and nonrhythmic compartments of the suprachiasmatic nucleus pacemaker. J Neurosci 21: 7742-7750.
  49. Ingram CD, Ciobanu R, Coculescu IL, Tanasescu R, Coculescu M, Mihai R. 1998 Vasopressin neurotransmission and the control of circadian rhythms in the suprachiasmatic nucleus. Prog Brain Res 119: 351-364.
  50. Jin X, Shearman LP, Weaver DR, Zylka MJ, deVries GJ, Reppert SM. 1999 A molecular mechanism regulating rhythmic output from the suprachiasmatic circadian clock. Cell 96: 57-68.
  51. Arima H, House SB, Gainer H, Aguilera G. 2002 Neuronal activity is required for the circadian rhythm of vasopressin gene transcription in the suprachiasmatic nucleus in vitro. Endocrinology 143: 4165-4171.
  52. Romijin HJ, Sluiter AA, Pool CW, Wortel J, Buijs RM. 1997 Evidence from confocal fluorescence microscopy for a dense, reciprocal innervation between AVP-, somatostatin-, VIP/peptide histidine isoleucine-, GRP, and peptide histidine isoleucine/GRP-immunoreactiave neurons in the rat suprachiasmatic nucleus. Eu J Neurosci 9: 2613-2623.
  53. Gillette MU, Tischkau SA. 1999 Suprachiasmatic nucleus: The brain’s circadian clock. Recent Prog Brain Res 54: 33-59.
  54. Moore RY.1996 Neural control of the pineal gland.  Behav Brain Res 73:  125-130.
  55. Moore RY. 1996 Entrainment pathways and the functional organization of the circadian system.Prog. Brain Res. 111: 103-119..
  56. Arendt J.2000 Melatonin, circadian rhythms, and sleep. N Engl J Med. 343: 1114-1116.
  57. Karsch FJ, Bittman EL, Robinson JE, Yellon SM, Wayne NL, Olster DH, Kaynard AH. 1986 Melatonin and photorefractoriness: loss of response to the melatonin signal leads to seasonal reproductive transitions in the ewe. Biol Reprod. 34: 265-274.
  58. Azzali G, Arcari ML, Cacchini A, Toni R.2003 Fine structure and photoperiodical seasonal changes in Pars tuberalis of hibernating bats.  It J Anat Embriol 108(1):49-64.
  59. Drew KL, Buck CL, Barnes BM, Christian SL, Rasley BT, Harris MB. 2007 Central nervous system regulation of mammalian hibernation:implications for metabolic suppression and ischemia tolerance.  J Neurochem 102: 1713-1726.
  60. Kalsbeek A., Buijs RM.2002 Output pathways of the mammalian suprachiasmatic nucleus: coding circadian time by transmitter selection and specific targeting.  Cell Tissue Res 309:109–118.
  61. Feillet CA.2010 Food for thoughts: feeding time and hormonal secretion.  Journal of Neuroendocrinology 22, 620–628.
  62. Girotti, M., Weinberg, M.S., Spencer, R.L. 2009. Diurnal expression of functional and clock-related genes throughout the rat HPA axis: system-wide shifts in response to a restricted feeding schedule. Am J Physiol Endocrinol Metab 296, 888–897.
  63. Fuller MP, Gooley JJ, Saper CB. 2006 Neurobiology of the sleep-wake cycle:sleep architecture, circadian regulation and regulatory feedback.  J Biol Rythms 21: 482-493.
  64. Saper CB, Chou TC, Scammell TE. 2001 The sleep switch:hypothalamic control of sleep and wakefulness.  Trend Neurosci 24: 727-731.
  65. Sherin JE, Shiromani PJ, McCraly RW, Saper CB. 1992 Activation of ventrolateral preoptic neurons during sleep.Science 271, 216-219, 1996.
  66. Lugaresi E.1992 The thalamus and insomnia.  Neurology 42: 28-33.
  67. Steriade M.1994 The thalamus and sleep disturbances.  In:  Guilleminault C, Lugaresi E, Montagna P, Gambetti P, eds, Fatal Familial Insomnia:  Inherited Prion Diseases, Sleep and the Thalamus.  New York, Raven Press, pp. 177-189.
  68. Moruzzi G, Magoun HW.1949 Brain stem reticular formation and activation of EEG.  Electroecaphalogry Clin Neurol 1: 455-473.
  69. Toni R, Malaguti A, Benfenati F, Martini L. 2004 The human hypothalamus: a morphofunctional perspective.J Endocrinol Invest 27 (supp to n.6):  73-94.
  70. Sherin JE, Elmquist JK, Torrealba F, Saper CB. 1998 Innervation of histaminergic tuberomammillary neurons by GABAergic and galaninergic neuros in the ventrolateral preoptic nucleus of the rat.J Neurosci 18: 4705-4721.
  71. Ericson H,Blomqvist A, Köhler C. 1991 Origin of neuronal inpus to the region of 6he tuberomammliary nucleus of the rat brain.  J Comp Neurol 311: 45-64.
  72. Yoshimoto Y, Sakai K, Luppi PH, Fort P, Salvert D, Jouvet M. 1989 Forebrain afferents to the cat posterior hypothalamus:a double labelling study.  Brain Res Bull 23: 83-104.
  73. Gervasoni D, Peyron C, Rampon C, Barbagli B, Chouvet G, Urbain N, Fort P, Luppi PH. 2000 Role and origin of the GABAergic innervation of dorsal raphé serotoninergic neurons. J Neurosci 20: 4217-4225.
  74. Steininger TL, Gong H, McGinty D, Szymusiak R. 2001 Subregional organization of preoptic area/anterior hypothalamic projections to arousal-related monaminergic cell groups.J Comp Neurol 429: 638-653.
  75. Parent A, Steriade M. 1984 Midbrain tegmental projections of nucleus reticularis thalami of cat and monkey:a retrograde transport and antidromic invasion study.  J Comp Neurol 229: 548-558.
  76. Saper CB. 2000 Hypothalamic connections with the cerebral cortex.Prog Brain Res 126: 40-38.
  77. Fuller PM, Sherman D, Pedersen NP, Saper CB, Lu J. 2011 Reassessment of the structural basis of the ascending arousal system. J Comp Neurol. 519: 933–956.
  78. Saper CB, Scammell TE, Lu J. 2005 Hypothalamic regulation of sleep and circadian rhythms.Nature 437: 1257-1263.
  79. Buzsaki G, Bickford RG, Ponomareff G, Thal LJ, Mandel R, Gage FH. 1988 Nucleus basalis and thalamic control of neocortical activity in the freely moving rat.J Neurosci 8: 4007-4026.
  80. Kampfl A, Franz G, Aichner F, Pfausler B, Haring HP, Felber S, Luz G, Schocke M, Schmutzhard E.1998 The persistent vegetative state after closed head injury: clinical and magnetic resonance imaging findings in 42 patients. J Neurosurg. 88:809-816.
  81. Manford M, Andermann F. 1988 Complex visual hallucinations. Clinical and neurobiological insights. Brain.121:1819–1840.
  82. Aschkenasi CJ, Lee CE, Chemelli RM, Saper CB, Yanagisawa M, Elmquist JK. 2001 Differential expression of orexin receptors 1 and 2 in the rat brain.J Comp Neurol 435: 6-25.
  83. Trivedi P, Yu H, MacNeil DJ, Van der Ploeg LH, Guan XM. 1998 Distribution of orexin receptor mRNA in the rat brain.FEBS Lett 438: 71-75.
  84. Hagan JJ, Leslie RA, Patel S, Evans ML, Wattam TA, Holmes S, Benham CD, Taylor SG, Routledge C, Hemmati P, Munton RP, Ashmeade TE, Shah AS, Hatcher JP, Hatcher PD, Jones DN, Smith MI, Piper DC, Hunter AJ, Porter RA, Upton N. 1999 Orexin A activates locus coeruleus cell firin and increases arousal in the rat.Proc. Natl Acad Sci USA 96: 10911-10916.
  85. Brown RE, Sergeeva O, Eriksson KS, Haas HL. 2001 Orexin A excites serotoninergic neurons in the dorsal raphé nucleus of the rat.Neuroparmacology 40; 457-459.
  86. Methippara MM, Alam MN, Szymusiak R, McGinty D. 2000 Effects of lateral preoptic area application of orexin-A on sleep-wakefulness.NeuroReport 11: 3423-3426.
  87. Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa M. 1999 Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation.Cell 98: 437-451.
  88. Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. 1999 The sleep disorder canine narcolepsy is caused by a mutation of the hypocretin (orexin) receptor 2.Cell 98: 365-376.
  89. Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. 2000 Hypocretin (orexin) deficiency in human narcolepsy.Lancet 355: 39-40.
  90. Kocsis B, Kaminski M. 2006 Dynamic changes in the direction of theta rhythmic drive between supramammillary nucleus and the septohippocampal system.Hippocampus 16: 531-540.
  91. Lu J, Sherman D, Devor M, Saper CB. 2006 A putative flip-flop switch for control of REM sleep.Nature 441: 589-594
  92. Lugaresi E, Montagna P. 1994 Thalamus, sleep and circadian funtions.In:  Guilleminault C, Lugaresi E, Montagna P, Gambetti P, eds, Fatal Familial Insomnia:  Inherited Prion Diseases, Sleep and the Thalamus.  New York, Raven Press, pp 215-219.
  93. Marini G, Imeri L, Mancia M. 1988 Changes in sleep-waking cycle induced by lesions of medial dorsalis thalamic nuclei in the cat.Neurosci Lett 85: 223-227.
  94. Lugaresi E, Medori R, Montagna P, Baruzzi A, Cortelli P, Lugaresi A, Tinuper P, Zucconi M, Gambetti P. 1986 Fatal familial insomnia and dysautonomia with selective degeneration of thalamic nuclei.New Engl J Med 315: 997-1003.
  95. Lu J, Shang YH, Chou T, Gauss SE, Elmquist JK, Shiromani P, Saper CB.2001 Contrasting effects of ibotenate lesions of the paraventricular nucleus and subparaventricular zone on sleep-wake cycle and temperature regulation.  J Neurosci 21: 4864-487.
  96. Gooley JJ, Schomer A, Saper CB. 2006 The dorsomedial hypothalamic nucleus is critical for the expression of food-entrainable circadian rhythms.Nat Neurosci 9: 398-407.
  97. Chou TC, Scammell TE, Gooley JJ, Gaus SE, Saper CB, Lu J. 2003 Critical role of dorsomedial hypothalamic nucleus in a wide range of behaviorl circadian rhythms.J Neurosci 23: 10691-10702.
  98. Saper CB, Lu J, Chou TC, Gooley J. 2005 The hypothalamic integrator of circadian rhythms.Trends Neurosci 28: 152-157.
  99. Harrington ME. 1997 The ventral lateral geniculate nucleus and the intergeniculate leaflet: interrelated structures in the visual and circadian systems.Neurosci Biobehav Rev 21; 705-727.
  100. Donlea JM, Thimgan MS, Suzuki Y, Gottschalk L, Shaw PJ. 2011Inducing sleep by remote control facilitates memory consolidation in Drosophila. Science. 332:1571-1576.
  101. Elbaz I, Foulkes NS,Gothilf Y, Lior Appelbaum L. 2013Circadian clocks, rhythmic synaptic plasticity and the sleep-wake cycle in zebrafish.Frontiers in Neural Circuits 7: 1-7.  
  102. Saper CB, Fuller PM, Pedersen NP, Lu J, Scammell TE. 2010Sleep state switching. Neuron. 68:1023-1042.
  103. Nofzinger EA, Buysse DJ, Germain A, Price JC, Miewald JM, Kupfer DJ. 2004Functional neuroimaging evidence for hyperarousal in insomnia. Am J Psychiatry. 161:2126–2128.
  104. Porkka-Heiskanen T, Strecker RE, Thakkar N, Bjiorkum AA, Greene RW, McCarley RW. 1997 Adenosine:a mediator of sleep-inducing effects of prolonged wakefulness.  Science 276, 1265-1268.
  105. Shilber U. 2007 The daily timing of gene expression and physiology in mammals.Dialogues Clin Neurosci 9: 257-272.
  106. Naylor E, Bergman BM, Krauski K, Zee PC, Takahashi JS, Vitaterna MH, Turek FW.2000 The circadian clock mutation alters sleep homeostasis in the mouse.  J Neurosci 20: 8138-8143.
  107. Wisor JP, O’Hara BF, Terao A, Selby CP, Kilduff TS, Sancar A, Edgar DM, Franken P.2002 A role for cryptochromes in sleep regulation.  BMC Neurosci 3: 20.
  108. Lapolsky A, Easton A, Dugovic C, Walisser J, Bradfield C, Turek F. 2005 Deletion of the mammalian circadian clock gene Bmal1/Mop3 alters baseline sleep architecture and the response to sleep deprivation.Sleep 2: 395-409.

622  .Sahar S, Sassone-Corsi P. 2012  Regulation of metabolism: the circadian clock dictates the time. Trends Endocrinol Metab. 3: 1–8.

Fetal and Neonatal Sterol Metabolism

ABSTRACT

 

Cholesterol is critical during the development of embryos, fetuses and neonates to support their growth and development. Cholesterol is a structural component of membranes in every cell, it is involved with numerous signaling events, and it is the precursor for key steroid hormones.  All individuals, either in utero or post-partum, have two sources of cholesterol, endogenous and exogenous. In the embryo and fetus, endogenous cholesterol comes from de novo synthesis and exogenous sources originate in the maternal circulation; maternal cholesterol-carrying lipoproteins are taken up from the maternal circulation by the placenta or yolk sac, processed, and transported across cells to the embryo or fetus. In the neonate, endogenous cholesterol is also synthesized de novo whereas exogenous cholesterol is derived from the diet. Changes in maternal metabolism (diabetes or obesity) or adverse pregnancy outcomes (preterm births or preeclampsia) could lead to altered fetal sterol metabolism. In this review, we will examine fetal and neonatal cholesterol metabolism in complicated and uncomplicated pregnancies. Early identification of neonatal cholesterol abnormalities could identify infants in need of immediate treatments, mostly due to genetic disorders, and infants that could be at long-term risk of metabolic diseases.

 

SOURCES OF FETAL CHOLESTEROL 

 

A significant amount of cholesterol is accrued during gestation.  A newborn that weighs ≈4.5 kg requires ≈12 g of cholesterol with concentrations ranging from ≈2.2 mg cholesterol/g liver and peripheral tissues and ≈8 mg cholesterol/g neuronal tissues [reviewed in (1,2)].  Cholesterol is not only needed to maintain structural integrity but is also required for a variety of signaling events and as precursor of steroid hormones. Signaling that depends on cholesterol is varied, and includes the presence of cholesterol in specific regions of the membranes (lipid rafts) to allow signaling proteins to aggregate and bind specific scaffold proteins and to form endosomes (3-7), the formation of unique covalent bonds between cholesterol and Hedgehog (HH) and Smoothened (8,9), and the conversion of cholesterol to active oxysterols (10,11). The fetus has two sources of cholesterol, that synthesized de novo and that obtained from the maternal circulation. 

 

Sterol Synthesis 

 

Sterol synthesis rates are much greater in the fetus than in the adult in several species (2,12-16), including humans (2).  Rates are high enough to account for a significant proportion of the fetal cholesterol in rodents (12,17-19).  Synthesis rates vary between different fetal tissues and is greatest in the liver early in gestation.  As gestation progresses, hepatic synthesis decreases to rates similar to other tissues by late in gestation (13).  While the brain has the greatest cholesterol concentration, synthesis rates are not extremely elevated as cholesterol is turned over at very low rates in the brain (1). 

 

Sterol biosynthesis is regulated at several different steps in the biosynthetic pathway primarily by the processing of transcription factor sterol regulatory element-binding protein-2 (SREBP-2) to the mature form. When cellular cholesterol concentrations are elevated in adult tissues, sterol synthesis rates and mature SREBP-2 levels are decreased (14,20). In contrast, when cellular cholesterol concentrations are elevated in the fetus, sterol synthesis rates are suppressed only marginally and mature SREBP-2 levels do not decrease (14), suggesting constitutive processing of SREBP, and higher synthesis rates in the fetus. This same lack of regulation in fetal tissues was found when fetal hepatocytes were treated with lipoprotein-cholesterol (21) and when fetuses were exposed to polyunsaturated fatty acids in vivo (22). Interestingly, sterol synthesis rates can be stimulated in vitro by hormones synthesized by the placenta, including estrogens and progesterone, possibly to ensure that essential lipids are abundantly present (23). 

 

Maternally-Derived Cholesterol

 

The second source of fetal cholesterol originates in the maternal circulation. Several lines of evidence support the presence of maternally-derived cholesterol in the fetal circulation. First, while there is no correlation between maternal and newborn cholesterol concentrations in term or late preterm infants, there is a direct relationship between maternal and fetal plasma cholesterol concentrations early in gestation (24).  Second, correlations between maternal and fetal concentrations occur when maternal plasma concentrations are correlated to fetal arterial and not fetal venous plasma concentrations (25). Third, fetuses of mothers with higher plasma cholesterol levels have increased intimal plaque (24).  Fourth, there are significant amounts of plant sterols in the newborn circulation, 40-50% of that found in the maternal circulation (26).  As these sterols are only obtained from the diet of the mother, they must cross the placental barrier. Fifth, fetuses lacking the ability to synthesize cholesterol due to a null/null mutation in one of the enzymes of the cholesterol biosynthetic pathway, such as dehydrocholesterol-7 reductase, have measurable, though low, amounts of cholesterol at birth (27,28).  Finally, using a 4-vessel sampling method in pregnant women, researchers measured substantial uptake of cholesterol by the fetus with more maternal HDL-C being taken up by the fetus vs maternal LDL-C (29). 

 

The route by which maternally-derived cholesterol is delivered to the fetus differs as the maternal-fetal interface changes during gestation (see Figure 1).  Very early in gestation (≈first 5 weeks), endocrine gland secretions containing maternally-derived cholesterol as lipid droplets bathe the blastocyst as they invade the uterine wall and are the main source of maternally-derived lipids, and overall histotrophic nutrition (30).  As gestation progresses (≈5th to ≈10th week of gestation), the newly formed secondary yolk sac of the embryo floats in the nutrient-rich exocoleom cavity (31) (Fig. 1A).  Nutrition at this stage is still primarily histotrophic and consists primarily of lipid-containing secretions from uterine glands and possibly some maternal lipoproteins from maternal blood which has seeped into the exocoleomic cavity (30). The human yolk sacs are not inverted, as they are in rodents, such that the highly absorptive apical side of the yolk sac faces inward (30-32), and the lipids would need to enter the yolk sac cavity via lipoprotein or endocytic receptors or other carriers (33-36).  Once taken up, the cholesterol from the lipids or lipoproteins (37) are repackaged into nascent lipoproteins (34,38,39), which are secreted into the vitelline duct artery which is integrated into the midgut of the embryo (30,31,40).  In rodents, an inability to form lipoproteins in the yolk sac is lethal (41).

Figure 1. Scheme of the sources of cholesterol from different times of gestation. A. From about 5-10 weeks of gestation, the primary source of nutrition for the embryo/fetus is from uterine gland secretions in the form of lipid droplets (dark blue circles). There is a small amount of maternal lipoproteins (green and orange circles) that is also present in the exocoleum cavity from leakage from spiral arteries. The lipids diffuse into the yolk sac cavity and are taken up by the apical side of the yolk sac’s endoderm cells via receptor- and receptor-independent mechanisms. The cholesterol is released from other components in lysosomes and repackaged into APOB-containing lipoproteins (and perhaps other lipoproteins) and secreted into the vitteline vessels which combine with the fetal circulation in the mid-gut. B. From 10 weeks of gestation to parturition, the fetus obtains its cholesterol from the maternal circulation in the placenta. The maternal cholesterol-carrying lipoproteins are taken up by the apical side of multi-nucleated syncytialized trophoblasts, is released from other components of the lipoproteins in lysosomes, and transported to the basolateral side of the trophoblasts. The cholesterol exits the trophoblasts to the stroma or cells within the stroma, is taken up by the fetal endothelial cells, is processed or transported to the opposite side where the cholesterol exits the cells. The routes of exit from the trophoblasts or endothelial cells are discussed in Figure 2.

As gestation progresses to the second and third trimesters, nutrition becomes hemotrophic meaning nutrition is obtained from maternal blood.  Early in gestation, the placenta does not transport nutrients as the spiral arteries that supply the placenta with maternal lipoprotein-containing blood are “plugged” by extravillous trophoblasts, blocking maternal blood from entering placental spaces that surround the trophoblasts (reviewed in (42,43)).  At about 10 weeks of gestation, the “plugs” disintegrate, allowing maternal blood to enter the intervillous spaces of the placenta, directly bathing the syncytialized trophoblasts (Fig. 1B); the syncytiotrophoblasts of the placenta are polarized and take up nutrients from the maternal circulation from its apical side. The maternal blood within the intervillous space that bathes the trophoblasts exchanges 3-4 times each minute, making this an excellent source of nutrients.  The maternally-derived lipoproteins are taken up via receptor-independent and receptor-dependent processes, including the LDL receptor, the VLDL receptor, the class A scavenger receptor, the LDL receptor-related protein (LRP), the APOE receptor 2, megalin, cubilin, and the scavenger receptor class B type I (SR-BI) (35,42).  The sterol-containing lipoproteins are then transported to lysosomes where the sterol is released from lipoproteins via numerous lysosomal hydrolases and transported to the basolateral side of the trophoblasts by carrier and transport proteins (37).  On the basolateral (fetal-facing) side of trophoblasts, the sterols exit the trophoblasts into the stroma, are subsequently taken up by and cross fetal endothelial cells, and ultimately exit these cells and enter the fetal circulation. It also is possible that the LDL or HDL are transcytosed across trophoblasts and endothelial cells as whole particles (44,45).  

 

There are several routes by which cholesterol can exit the basolateral side of trophoblasts and fetal endothelial cells (Figure 2), including secretion of particles and efflux of sterol to acceptors.  Both processes can be regulated at several points.  First, human placentas and cultured cells secrete newly synthesized APOB-containing lipoproteins (46,47) and APOA1 and APOE (48).  As in other cells that secrete lipoproteins (hepatocytes and enterocytes), cellular cholesterol can drive lipoprotein-cholesterol secretion from trophoblasts (49).  It is likely that other substrates would increase lipoprotein secretion from these cells.  Estradiol, which is elevated during pregnancy, also increases secretion of nascent APOB-containing lipoproteins from cultured trophoblasts (47).  Second, cholesterol can be effluxed from cells by either aqueous diffusion or by ATP binding cassette subfamily A member 1 (ABCA1), ABCG1, or SR-BI, all proteins which are expressed in trophoblasts (42,50).  Regulation of efflux occurs at the level of cellular proteins that mediate efflux, including SR-BI, ABCA1, and ABCG1 (50-54), and by the level and type of acceptor in the circulation (55).  The regulatory protein, liver X receptor (LXR), is a key mediator of ABCA1 and ABCG1 (56) (57). The levels of LXR are enhanced by oxysterols (58).  Thus, changes in cellular oxysterols can enhance efflux via ABCA1/G1 (see Adverse pregnancy outcomes).  The movement of cholesterol by SR-BI is regulated by cholesterol concentrations and phospholipids in the cells and in the accepting HDL (59).  The expression of SR-BI also is regulated by a number of factors, including cellular sterol levels (60).  Efflux is not only regulated by the proteins that enhance efflux, but also by the type and concentration of acceptors in endothelial spaces and circulation, with the amount of lipid-poor APOE or APOA1 and the composition of HDL being important.  For example, we found that lipid-poor HDL from a fetus with the Smith-Lemli-Opitz Syndrome (SLOS) that is unable to synthesize cholesterol is a better acceptor of trophoblast cholesterol than a non-SLOS fetal HDL (61).  Likewise, when HDL was changed to a better sterol-accepting particle by the phospholipid transfer protein, efflux from endothelial cells increased (62,63).  One other aspect of HDL which affects efflux is the proteome (64-66). Fetal HDL does contain more APOE than adult HDL, less cholesteryl ester transfer protein (CETP), and equal lecithin cholesteryl acyl transferase (LCAT),a combination of proteins which support a larger particle that can efflux more cholesterol via SR-BI and ABCG1 and can’t obtain cholesterol from other lipoproteins via CETP (67). 

 

Figure 2. Routes of exit of cholesterol from trophoblasts and fetal endothelial cells. Cholesterol exits these cells by being effluxed out of cells to acceptors in the plasma or by being secreted. There are several routes for efflux to occur, and all proteins involved have been found in the cell types listed; aqueous diffusion to acceptors (with lipid-poor acceptors being most efficient), SR-BI mediated, ABCG1-mediated, and ABCA1-mediated. In trophoblasts, studies have shown that cells can secrete lipoproteins and apolipoproteins which will carry sterols to the circulation as they exit the cell. Finally, exosomes carry cholesterol as they exit cells.

FETAL LIPOPROTEIN CHOLESTEROL CONCENTRATIONS AND COMPOSITION 

 

The functions of lipoproteins are to transport lipids through the plasma since cholesterol, and other lipids, are lipophilic and therefore not water soluble. The two lipoproteins that carry most of the circulating cholesterol are low-density lipoprotein (LDL) and high-density lipoprotein (HDL), with lower amounts being carried as very low-density lipoproteins (VLDL).  According to the National Health and Nutrition Examination Survey (NHANES), adults with an average age of 49±18 years have an average total cholesterol concentration of 193±42 mg/dl.  A majority of the plasma cholesterol in adults is carried as LDL (115±35 mg/dl) with HDL carrying less cholesterol (53±15 mg/dl), making an average LDL-C/HDL-C ratio in adults of 2.17 (68).  In contrast, plasma total cholesterol concentrations range from 51.4-96.8 mg/dl in term infants (69-80).  A greater proportion of cholesterol is carried as HDL (22.1-44.9 mg/dl) versus LDL (22.0-44.9 mg/dl) in the fetus compared to the adult leading to a ratio of LDL-C/HDL-C of 0.56-1.55 in the fetus/newborn, with an average ratio of 0.99 in term infants (69,71-76,79,81). 

 

Fetal plasma cholesterol concentrations are not constant throughout gestation, and most studies show concentrations to decrease as gestation progresses (75,82-84).  The biggest decreases appear to occur with LDL-C, possibly due to increased uptake of LDL by enhanced hepatic LDL receptor activity late in gestation (85).  Decreases are detected even when only term infants are compared by gestational ages, and decrease from ratios of 1.61 at 37-38 weeks of gestation to 1.27 at 41-42 weeks of gestation (83).  Not all studies measure a decrease in fetal plasma cholesterol with gestational age, such as a study in Korea (86), possibly due to the location of the study since most other studies were in resource-rich settings. 

 

Lipoproteins (HDL, LDL, VLDL) are not comprised of just one size and type of particle, but are comprised of a spectrum of sizes (subfractions) and subspecies that carry different proteins and have different functions (64-66).  This is especially true for HDL particles as over 250 distinct proteins have been associated with HDL with different combinations of protein leading to a myriad of functions (64-66,87).  Not surprisingly, fetal vs adult lipoproteins differ in composition and subfraction concentrations as well as total lipoprotein-cholesterol concentrations.  For example, fetal HDL particles are larger than adult HDL particles (88-90) and, small-dense LDL particles are more abundant in fetal compared to adult circulations (91).  In adults, the proteins carried by HDL are involved in oxidation, inflammation, hemostasis, vitamin transport, immunity, energy balance, and lipid transport (66,92).  In contrast, fetal HDL particles are enriched in proteins involved in coagulation and transport, and is lacking in proteins involved in anti-oxidative processes, such as paraoxonase I (PON1) (90).  The lack of PON1 on fetal HDL suggests that these particles do not have the same anti-oxidative capacity as that found in adults (90).  In addition, fetal HDL is enriched in APOE.  The excess APOE could enhance the uptake of fetal HDL into fetal tissues by members of the LDL receptor family, enhance efflux of cholesterol out of endothelial cells, and affect genes related to sterol metabolism and oxidation in fetal endothelial cells (93).  Unlike HDL, fetal VLDL and LDL compositions have not been studied in any detail as of yet.

 

REGULATION OF FETAL LIPOPROTEIN CHOLESTEROL CONCENTRATIONS

 

In the fetus as in the adult, plasma lipoprotein-cholesterol levels are regulated by the amount of cholesterol entering versus that exiting the circulation.  Adults in steady state have an equal amount of cholesterol entering and exiting the plasma unlike the fetus where the amount of cholesterol entering vs that exiting is not equal.  The lower levels of cholesterol in fetal plasma suggests less cholesterol entering the plasma or more exiting the plasma. 

 

Low-Density Lipoprotein (LDL) 

 

LDL-C originally enters the plasma after the liver synthesizes and secretes VLDL which is converted to LDL in the circulation.  Since the liver is not functionally developed in utero (94,95), lipoprotein production and secretion could be low, being at least part of the cause of the low fetal LDL-C levels.  The lower levels of circulating fetal cholesterol levels could also be due to an increase in uptake of lipoprotein cholesterol from the circulation by LDL receptors.  Using the in vivo catheterized pregnant sheep model, it was found that uptake of cholesterol by tissues is greater in utero than in the post-partum neonatal lamb (96).  This is not unexpected as fetal tissues require significant amounts of cholesterol for membrane formation and for steroid hormone synthesis (97-99). 

 

High-Density Lipoprotein (HDL) 

 

Interestingly, HDL-C levels are relatively elevated in the fetus.  Unlike VLDL and LDL, HDL is produced in the circulation and as such is not dependent upon the fetal liver for lipoprotein production. To produce HDL, cholesterol is effluxed from cells by lipid-poor APOA1 or APOE, followed by esterification of the cholesterol by lecithin cholesterol acyl transferase (LCAT), all of which are present in the fetal circulation (90).

 

ABNORMAL FETAL STEROL METABOLISM; IMPACT OF GENETIC ALTERATIONS AND ADVERSE PREGNANCY OUTCOMES   

 

Abnormal fetal sterol metabolism can come about by genetic alterations in the fetus and by influences of maternal factors (maternal obesity, diabetes, dyslipidemia, preeclampsia) on fetal metabolism. 

 

Genetic 

 

Even though two sources of cholesterol exist for the fetus, a majority of fetal cholesterol is likely derived from de novo synthesis.  Thus, changes in sterol synthesis could lead to unfavorable development.  There are several known genetic defects in the post-squalene cholesterol biosynthetic pathway that result in altered fetal phenotypes [reviewed in (50,100-102)]; one pre-squalene defect occurs (103).  Of these metabolic disorders, the most common is the SLOS.  Individuals with SLOS have affected midline facial features, multiple organ and limb malformations, and intellectual disability.  As sonic hedgehog (SHH) is expressed as early as 3 weeks after fertilization, and SHH is essential in a number of key developmental processes (104,105), changes in activation could have very early and significant effects.  Indeed, lower SHH signaling has been associated with altered signaling that occurs in individuals with SLOS (106).  Lower sterol synthesis rates in individuals with SLOS could also lead to reduced growth rates and intrauterine growth retardation (IUGR) (107).  Though it was originally thought that the syndrome was due to a lack of cholesterol, the accumulation of 7-dehydrocholesterol (DHC) likely plays a role in the progression of the disease as well (108). Interestingly, a large percentage of individuals with SLOS are autistic (109) and some individuals with autism have been shown to have altered cholesterol metabolism (110) and dyslipidemia (111,112).  Disruption of enzymes that occur pre-mevalonate in the sterol biosynthesis pathway has not been documented in live newborns, and are associated with embryonic lethality in murine deletion models [reviewed in (50)]. 

 

Adverse Pregnancy Outcomes 

 

Pregnancies complicated with diabetes, obesity, or preeclampsia often have adverse outcomes, including preterm births, altered growth rates, and in the most severe cases, stillbirths and infant mortality. The link between the altered metabolism in the pregnant females and the fetuses are often unknown but are hypothesized to be related to inflammation or oxidative stress within the placenta or fetus. Oxidative stress increases in pregnancy, and especially in pregnancies associated with diabetes, obesity, and preeclampsia (113,114).  There is an increase in oxygen species during oxidative stress which are involved in the conversion of cholesterol to oxysterols (115,116). 

 

Placenta and Fetal Endothelial Cells

 

Oxysterols are detrimental during development as these cholesterol derivatives affect a number of signaling pathways including activation of the liver X receptor (LXR) (117) and inhibition of hedgehog signaling (118).  The oxysterol-induced increase in LXR activation enhances cholesterol and oxysterol efflux from these cell types.  Indeed, fetal endothelial cells of women with gestational diabetes had increases in LXR target genes, ABCA1 and ABCG1, and increased cholesterol efflux (119).  Likewise, HDL-mediated cholesterol efflux and placental 27-hydroxycholesterol were increased in women with preeclampsia (120). 

 

Studies have shown reduced HDL-C and elevated LDL-C levels in preeclamptic infants (121) and increased oxidative modifications of LDL and HDL associated with decreased PON1 activity (122).  Changes in the subfraction concentrations have been detected, as well, and infants of women with type I diabetes had greater HDL2-C and -phospholipid concentrations vs women without diabetes (123). 

 

These changes in sterol metabolism in the placenta, endothelial cells and fetus can lead to a variety of outcomes ranging from beneficial to adverse.  First, an increase in efflux due to increased placental oxysterol levels would increase fetal cholesterol and oxysterol levels, which could be deleterious to the fetus as oxysterols can inhibit hedgehog signaling (118).  In contrast, this same increase in efflux from cells with increased oxysterol could reduce the sterol concentrations in the placenta, possibly protecting the cells from a build-up of oxysterols, which could be improve placental metabolism. 

 

NEONATAL LIPOPROTEIN CHOLESTEROL CONCENTRATIONS AND CHOLESTEROL METABOLISM

 

The three major sources of nutrition in the United States during neonatal and early infancy are human milk, cow milk-based formulas, and soy milk-based formulas. The composition of these types of diet differs in several factors that may theoretically influence cholesterol homeostasis including cholesterol content, polyunsaturated/saturated fatty acid ratio (P/S ratio), protein composition, phytoestrogen content, and the presence of hormones specific to breast milk.  More recent components of milk which also can affect metabolism include miRNAs, prebiotics, and extracellular vesicles (124-126). 

 

As with the fetus, neonates are in a rapid growth phase and require significant cholesterol for growth, energy, and normal cellular function. Infants fed human milk receive much greater quantities of cholesterol than those fed commercial formulas. Human milk contains between 10-15 mg/dl of cholesterol, cow milk-based formulas contain 1-4 mg/dl of cholesterol, and soy -based formulas contain no cholesterol. The soy-based formulas contain phytosterols (plant sterols), which actually inhibit cholesterol absorption (127).  Not unexpectedly, breast-fed infants have higher serum cholesterol concentrations compared to formula-fed infants (128,129). In contrast to the fetus which does not suppress sterol synthesis rates, rapidly-growing neonates do suppress sterol synthesis (130-132). 

 

LONG-TERM IMPACT OF ALTERED FETAL AND NEONATAL STEROL METABOLISM    

 

In the early 1990s, Dr. David Barker discovered that persons growing up in less affluent areas of England and Wales were at an increased risk for infant mortality and long-term ischemic heart disease compared to persons in more affluent areas (133).  Specifically, the adverse long-term consequences of heart disease were related to low birthweights. This relationship was confirmed by others [reviewed in (134,135)], and was expanded to also include infants who are born large for gestational age (LGA), forming a U-shaped curve.  Because of his early seminal work in this area, the “programming” of metabolism by early life environment has been coined the “Barker hypothesis” or DOHaD (Developmental Origins of Health and Disease). The importance of early nutrition has been labeled “1000 days” as that is the time from conception to the second birthday when much growth and development (and programming) (136).

 

It is difficult to identify any long-term effects that are specific to cholesterol as changes in cholesterol levels in the newborn or in utero are often associated with the oxidative stress which accompanies the adverse outcomes it is associated with.  For example, some studies have shown infants of preeclamptic mothers or preterm infants to be at an increased risk of heart disease later in life (137-143).  The long-term changes in metabolism that lead to programming in adulthood are likely epigenetic changes in genes controlling metabolism [reviewed in (144)].  Indeed, the greatest epigenetic activity occurs in the first 1000 days of life (145).  There are some recent treatments that are directed at changing the epigenome postnatally (not for newborns or neonates), including statins which are proposed to modify histones and various dietary regimes which can affect methylation status (146), and prenatally, including anti-oxidant compounds to reverse programming (147).

 

It is not only the in utero environment which has the potential to lead to programming of metabolic disease or heart disease, but also the type of diet fed to the newborn (148).  Since breast milk and formulas vary more than just in their cholesterol content, it is almost impossible to determine if early life cholesterol due to consumption of breast milk vs soy-based formulas affects age-related development of heart disease due to so many other non-sterol-based factors.  However, if one were to focus solely on cholesterol, one hypothesis would be that Infants fed cholesterol-containing human milk could be protective.  Support for this is that adult men and women who were breast-fed in infancy had lower serum cholesterol concentrations (149) or higher HDL-C levels (150) compared to adults who were not fed breastfed; BMI was also lower in adults that had been breast-fed (150).  Likewise, plasma total cholesterol was significantly higher in adult males that were breast fed for the shortest period of time when compared to those who were breast fed for longer times (151).  In contrast, plasma cholesterol concentrations in children and baboons fed either breast milk or formula had either no difference in plasma cholesterol levels or lower plasma cholesterol levels after being fed formula (152).  A review of the literature suggests that the differences in studies were due to studies using exclusive breastmilk versus those using both breast milk and formula (153), plus other outcomes could be important, including intellect and BMI (154).  Future studies are needed to better characterize the long-term effects of early cholesterol exposure on cholesterol metabolism in later childhood and adulthood.

 

CLINICAL SIGNIFICANCE OF FTAL AND NEWBORN PLASMA CHOLESTEROL CONCENTRATIONS

 

There are a few definitive clinical identifications that can be diagnosed with early life plasma cholesterol concentrations.  As discussed earlier, plasma cholesterol levels are lower in the newborn and quite variable, making it difficult to identify infants at risk of becoming hypercholesterolemic, even newborns that are heterozygous for familial hypercholesterolemia (155).  By one year of age, however, plasma levels are more stable and hypercholesterolemia becomes more obvious (155), taking into account the consumption of cholesterol-containing breastmilk at that time. This might be especially important in infants of women with altered metabolic conditions associated with oxidative stress.  The problem is that these cholesterol levels are either not routinely measured or not reported.

 

The plasma sterol concentrations, however, can be used to define various genetic disorders.  As discussed previously, there are known defects in the post-squalene cholesterol biosynthetic pathway that result in altered fetal phenotypes [reviewed in (50,100-102)], each with unique sterol compositions depending on where the defect in the sterol biosynthetic pathway occurs.  Thus, these syndromes can be identified by assaying for the specific sterols (see Table 1).  This is especially useful in the milder phenotypes that might be missed, especially SLOS due to its mild form and higher prevalence. Assays for these dehydrocholesterols must be done by gas chromatography to measure non-cholesterol sterol levels, not the commonly used enzymatic assay which measures sterol levels and not type. 

 

Table 1. Disorders of Cholesterol Biosynthesis*

Disease

Inheritance

Gene Defect

Laboratory Findings

Phenotype

Smith-Lemli-Optiz syndrome

AR

7-dehydrocholesterol reductase gene (DHCR7)

Elevated 7- dehydrocholesterol (DHC) and 8-DHC levels

 

Characteristic craniofacial appearance (i.e., ptosis, small upturned nose, and micrognathia, cleft palate); microcephaly; limb anomalies (proximally placed thumbs, polydactyly, and 2–3 toe syndactyly); slow growth and poor weight gain; potential cardiac and gastrointestinal anomalies and intellectual disability; severity depends on mutation (null to leaky)

Sterol-4-demethylase complex

AR

SC4MOL gene defect

Elevated 4,4’-demethyl- & 4-monomethyl-sterols

Microcephaly; cataracts; slow grow; dermatitis (scaling, erythroderma)

Desmosterolosis

AR

DHCR24

Very rare with only a few patients reported.

Elevated desmosterol

Craniofacial (dysmorphic features, i.e., micro/macrocephaly, cleft palate), ambiguous genitalia; short limbs and osteosclerosis; slow growth

Lathosterolosis

AR

Lathosterol 5-desaturase (SC5D)

Very rare with only a few patients reported.

Elevated lathosterol

 

SLOS like phenotype; craniofacial (subtle dysmorphic features, i.e., microcephaly, upturned nose); micrognathia; ptosis; cataracts; polysyndactyly or syndactyly; hypospadius

Chondrodysplasia punctata (Conradi-Hȕnermann syndrome; CDPX2)

X linked

Emopamil binding protein (EBP)

Elevated 8-DHC and 8(9)-cholestanol

 

Lethal in males

Females: craniofacial (asymmetric dysmorphic features); skin (generalized congenital ichthyosis on erythrothematous base), skeletal (stippling, rhizomelic limb shortening, scoliosis); ocular (cataracts); occasional malformations (cleft palate, hearing loss)

Congenital hemidysplasia with ichthyosiform erythroderma and limb defects (CHILD syndrome)

X linked

NADH steroid dehydrogenase-like (NSDHL) or EBP

Elevated 4-dimetyl, 4,4-dimethyl, and 4-carboxysterol intermediates (i.e., 4,4-dimethylcholesta-8, 24 dien-3β-ol)

 

Similar defects to CDPX2 but unilateral defects and no cataracts; lethal in males.

Females: striking unilateral distribution of anomalies. Generalized congenital ichthyosiform erythroderma and limb deformities (right >left). Internal malformations including CNS, renal and cardiac.

Hydrops-ectopic calcification-“moth eaten” skeletal dysplasia (HEM skeletal dysplasia, Greenberg dysplasia)

AR or AD

Lamin B receptor (LBR) with DHCR14 defect

Elevated 8(9), 14-dien- 3β-ol and cholesta-8(9),14,24-ien-3β-ol

Dysmorphic facial features, hydrops fetalis, cystic hydroma, lung abnormalities, severe short-limbed dwarfism with markedly disorganized cartilaginous and bony architecture (Moth eaten appearance of long bones)

Antley-Bixler syndrome

AR

CYP51A1-associated P450 cytochrome oxidoreductase (POR) gene

elevated levels of lanosterol and dihydrolanosterol

Craniosynostosis; choanal atresia; limb abnormalities (i.e., radio humeral synostosis, and femoral bowing); ambiguous genitalia

 

  *The disorders listed are post-squalene.  There is one defect in the pre-squalene pathway for cholesterol biosynthesis, Mevalonic Aciduria.

 

Though cholesterol levels are not often measured until 9-11 years of age per current recommendations for universal lipid screening per the American Academy of Pediatrics, earlier screening is appropriate if there is a strong family history of high cholesterol or early cardiovascular events. It should be noted infants associated with pregnancies complicated with adverse outcomes are at a higher risk to develop heart disease later in life (137-143,156-159).  Knowing this, interventions directed at improving cardiovascular risk, including maintaining a normal BMI, ideal blood pressure, ideal LDL-C etc., could be started earlier to prevent diseases from developing. 

 

SUMMARY

 

Cholesterol is essential for normal growth and development from the blastocyst through infancy. The cholesterol originates from an endogenous source (de novo synthesis) and an exogenous source (maternal lipoproteins and diet).  Due to its critical role in development, sterol synthesis rates are regulated less in the fetus than neonates.  If synthesis is reduced, possibly due to a genetic defect in the sterol biosynthetic pathway, abnormal development can occur.  Fetal cholesterol levels can be altered, including oxysterols, in pregnant women with various metabolic disorders, mostly those linked to oxidative stress.  The consequences of these changes are unknown because even though these infants are at an increased risk to develop age-related diseases later in life (130-136, 149-152), it is not known if the long-term effects are mediated by an early exposure to cholesterol/oxysterol or other factors associated with oxidative stress.  Regardless, offspring of mothers with hypercholesterolemia, preeclampsia, diabetes, obesity, or are born preterm should be monitored for future cardiovascular disease.      

 

ACKNOWLEDGEMENTS

 

We would like to acknowledge the contributions of the late James E. Heubi MD who was co-author on the original version of this chapter.

 

REFERENCES

 

  1. Dietschy JM, Turley SD. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J Lipid Res 2004; 45:1375-1397
  2. Dietschy JM, Turley SD, Spady DK. Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans. J Lipid Res 1993; 34:1637-1659
  3. Scorci-Thomas MG, Thomas MJ. Microdomains, inflammation, and atherosclerosis. Circ Res 2016; 118:679-691
  4. Lingwood D, Simons K. Lipid rafts as a membrane-organizing principle. Science 2010; 327:46-50
  5. Sheng R, Chen Y, Gee HY, Stec E, Melowic HR, Blatner NR, Tun MP, Kim YB, Kallberg M, Jujiwara TK, Hong JH, Kim KP, Lu H, Kusumi A, Lee MG, Cho W. Cholesterol modulates cell signaling and protein networking by specifically interacting with PDZ domain-containing scaffold proteins. Nat Comm 2012; 3:1249
  6. Lim C, Davis OB, Shin HR, Zhang J, Berdan CA, Jiang XC, Counihan JL, Ory DS, Nomura DK, Zoncu R. ER-lysosome contacts enable cholesterol sensing by mTOR1 and drive aberrant growth signalling in Niemann-Pick type C. Nat Cell Biol 2019; 21:1206-1218
  7. Anderson RH, Sochacki KA, Vuppula H, Scott BL, Bailey EM, Schultz MM, Kerkvliet JG, Taraska JW, Hoppe AD, Francis KR. Sterols lower energetic barriers of membrane bending and fission necessary for efficient clathrin-mediated endocytosis. Cell Rep 2021; 37:110008
  8. Porter JA, Young KE, Beachy PA. Cholesterol modification of hedgehog signaling proteins in animal development. Science 1996; 274:255-259
  9. Xiao X, Tang J-J, Peng C, Wang Y, Fu L, Qiu Z-P, Xiong Y, Yang L-F, Cui H-W, He X-L, Yin L, Qi W, Wong CCL, Zhao Y, Li B-L, Qiu W-W, Song B-L. Cholesterol modification of smoothened is required for hedgehog signaling. Mol Cell 2017; 66:154-162
  10. Torocsik D, Szanto A, Nagy L. Oxysterol signaling links cholesterol metabolism and inflammation via the liver X receptor in macrophages. Mol Aspects Med 2009; 30:134-152
  11. Griffiths WJ, Wang Y. Sterols, oxysterols, and accessible cholesterol:Signalling for homeostasis, in immunity and during development. Front Physiol 2021; 12:723224
  12. Haave NC, Innis SM. Cholesterol synthesis and accretion within various tissues of the fetal and neonatal rat. Metabolism 2001; 50:12-18
  13. Cavender CP, Turley SD, Dietschy JM. Sterol metabolism in fetal, newborn, and suckled lambs and their response to cholesterol after weaning. Am J Physiol 1995; 269:E331-E340
  14. Yao L, Jenkins K, Horn PS, Lichtenberg MH, Woollett LA. Inability to fully suppress sterol synthesis rates with exogenous sterol in embryonic and extraembyronic fetal tissues. Biochim Biophys Acta 2007; 1171:1372-1379
  15. Spady DK, Dietschy JM. Sterol synthesis in vivo in 18 tissues of the squirrel monkey, guinea pig, rabbit, hamster, and rat. J Lipid Res 1983; 24:303-315
  16. Rainey WE, Carr BR, Wang Z-N, Parker CRJ. Gene profiling of human fetal and adult adrenals. J Endocrinol2001; 171:209-215
  17. Belknap WM, Dietschy JM. Sterol synthesis and low density lipoprotein clearance in vivo in the pregnant rat, placenta, and fetus.Sources for tissue cholesterol during fetal development. J Clin Invest 1988; 82:2077-2085
  18. Woollett LA. Origin of cholesterol in the fetal Golden Syrian hamster: contribution of de novo sterol synthesis and maternal-derived lipoprotein cholesterol. J Lipid Res 1996; 37:1246-1257
  19. Jurevics HA, Kidwai FZ, Morell P. Sources of cholesterol during development of the rat fetus and fetal organs. J Lipid Res 1997; 38:723-733
  20. Shimomura I, Bashmakov Y, Shimano H, Horton JD, Goldstein JL, Brown MS. Cholesterol feeding reduces nuclear forms of sterol regulatory element binding proteins in hamster liver. Proc Natl Acad Sci USA 1997; 94:12354-12359
  21. Carr BR, Simpson ER. Cholesterol synthesis by human fetal hepatocytes:Effect of lipoproteins. Am J Obstet Gynecol 1984; 150:551-557
  22. Schmid KE, Woollett LA. Differential effects of polyunsaturated fatty acids on sterol synthesis rates in adult and fetal tissues of the hamster: Consequence of altered sterol balance. Am J Physiol 2003; 285:G796-G803
  23. Carr BR, Simpson ER. Cholesterol synthesis by human fetal hepatocytes:Effects of hormones. J Clin Endocrinol Metab 1984; 58:1111-1116
  24. Napoli C, D'Armiento FP, Mancini FP, Postiglione A, Wiztum JL, Palumbo G, Palinski W. Fatty streak formation occurs in human fetal aortas and is greatly enhanced by maternal hypercholesterolemia.Intimal accumulation of LDL and its oxidation precede monocyte recruitment into early atherosclerotic lesions. J Clin Invest 1997; 100:2680-2690
  25. Zhang RY, Dong S, Ma W, Cai X, Le Z, Xiao R, Zhou Q, Yu H. Modulation of cholesterol transport by maternal hypercholesterolemia in human full-term placenta. PLoS ONE 2017; 12:e1071934
  26. Vuorio AF, Miettinen TA, Turtola H, Oksanen H, Gylling H. Cholesterol metabolism in normal and heterozygous familial hypercholesterolemic newborns. J Lab Clin Med 2002; 140:35-42
  27. Linck LM, Hayflick SJ, Lin DS, Battalie KP, Ginat S, Burlingame T, Gibson KM, Honda M, Honda A, Salen G, Tint GS, Connor WE, Steiner RD. Fetal demise with Smith-Lemli-Opitz syndrome confirmed by tissue sterol analysis and the absence of measurable 7-dehydrocholesterol delta(7)-reductase activity in chorionic villi. Prenat Diagn 2000; 20:238-240
  28. Nowaczyk MJM, Farrell SA, Sirkin WL, Velsher L, Krakowiak PA, Waye JS, Porter FD. Smith-Lemli-Opitz (RHS) syndrome:Holoprosencephaly and homozygous IVS8-1G to C genotype. Am J Med Genet 2001; 103:75-80
  29. Horne H, Holme AM, Roland MCP, Holm MB, Haugen G, Henriksen T, Michelsen TM. Maternal-fetal cholesterol transfer in human term pregnancies. Placenta 2019; 87:23-29
  30. Hempstock J, Cindrova-Davies T, Jauniaux E, Burton GJ. Endometrial glands as a source of nutrients, growth factors and cytokines during the first trimester of human pregnancy: A morphological and immunohistochemical study. Reprod Biol Endocrinol 2004; 2:58-72
  31. Jauniaux E, Gulbis B. Fluid compartments of the embryonic environment. Hum Reprod Update 2000; 6:268-278
  32. Zohn IE, Sarkar AA. The visceral yolk sac endoderm provides for absorption of nutrients to the embryo during neurulation. Birth Defects Research A 2010; 88:593-600
  33. Jayalekshmi VS, Ramachandran S. Maternal cholesterol levels during gestation: boon or bane for the offspring? Mol Cell Biochem 2021; 476:401-416
  34. Lichtenberg MH, Wilke CS, McConihay JA, Granholm NA, Woollett LA. Yolk sac cholesteryl ester secretion rates can be manipulated in the Golden Syrian hamster:effect of yolk sac cholesterol concentrations. Biochim Biophys Acta 2005; 1735:214-221
  35. Wyne KL, Woollett LA. Transport of maternal LDL and HDL to the fetal membranes and placenta of the Golden Syrian hamster is mediated by receptor-dependent and receptor-independent processes. J Lipid Res 1998; 39:518-530
  36. Burke KA, Jauniaux E, Burton GJ, Cindrova-Davies T. Expression and immunolocalisation of the endocytic receptors megalin and cubilin in the human yolk sac and placenta across gestation. Placenta 2013; 34:1105-1109
  37. Meng Y, Heybrock S, Neculai D, Saftig P. Cholesterol handling in lysosomes and beyond. Trends Cell Biol2020; 30:452-466
  38. Shi W-K, Hopkins B, Thompson S, Heath JK, Luke BM, Graham CF. Synthesis of apolipoproteins, alphafoetoprotein, albumin, and transferrin by the human foetal yolk sack and other foetal organs. J Embryol Exp Morphol 1985; 85:191-206
  39. Cindrova-Davies T, Jauniaux E, Elliot MG, Gong S, Burton GJ, Charnock-Jones DS. RNA-seq reveals conservation of function among the yolk sacs of human, mouse, and chicken. Proc Natl Acad Sci 2017; 114:E4753-E4761
  40. Ross C, Boroviak TE. Origin and function of the yolk sac in primate embryogenesis. Nat Comm 2020; 11:3760
  41. Farese RV, Jr., Cases S, Ruland SL, Kayden HJ, Wong JS, Young SG, Hamilton RL. A novel function for apolipoprotein B:lipoprotein synthesis in the yolk sac is critical for maternal-fetal lipid transport in mice. J Lipid Res 1996; 37:347-360
  42. Woollett LA, Catov JM, Jones HN. Roles of maternal HDL during pregnancy. Biochim Biophys Acta - Mol Cell Biol Lipids 2022; 1867:159106
  43. Weiss G, Sundl M, Glasner A, Huppertz B, Moser G. The trophoblast plug during early pregnancy: a deeper insight. Histochem Cell Biol 2016; 146:749-756
  44. Cavelier C, Rohrer L, von Eckardstein A. ATP-Binding cassette transporter A1 modulates apolipoprotein A-I transcytosis through aortic endothelial cells. Circ Res 2006; 99:1060-1066
  45. Armstrong SM, Suglyama MG, Fung KY, Gao Y, Want C-N, Levy AS, Azizi P, Roufael M, Zhu SN, Neculai D, Yin C, Bolz SS, Seldah NG, Cybulsky M, Helt B, Lee WW. A novel assay uncovers an unexpected role for SR-BI in LDL transcytosis. Cardiovasc Res 2015; 108:268-277
  46. Madsen EM, Lindegaard MLS, Andersen CB, Damm Pl, Nielsen LB. Human placenta secretes apolipoprotein B-100-containing lipoproteins. J Biol Chem 2004; 279:55271-55276
  47. Kamper M, Mittemayer F, Cabuk R, Gelles K, Ellinger I, Hermann M. Estrogen-enhanced apical and basolateral secretion of apolipoprotein B-100 by polarized trophoblast-derived BeWo cells. Biochimie 2017; 138:116-123
  48. Melhem H, Kallol S, Huang X, Luthi M, Ontsouka CE, Keogh A, Stroka D, Thormann W, Schneider H, Albrecht C. Placental secretion of apolipoprotein A1 and E: the anti-atherogenic impact of the placenta. Sci Rep 2019; 9:6225
  49. Schmid KE, Davidson WS, Myatt L, Woollett LA. The transport of cholesterol across a BeWo cell monolayer: Implications for net transport of sterol from the maternal to fetal circulation. J Lipid Res 2003; 44:1909-1918
  50. Woollett LA. Where does fetal and embryonic cholesterol originate and what does it do? Ann Rev Nutr 2008; 28:97-114
  51. Baardman ME, Kerstjens-Frederikse WS, Berger MK, Hofstra RM, Plosch T. The role of maternal-fetal cholesterol transport in early fetal life: current insights. Biol Reprod 2013; 88:24
  52. Lindegaard ML, Wassif CA, Varisman B, Amar M, Wasmuth EV, Shamburek R, Nielsen LB, Remaley AT, Porter FD. Characterization of placental cholesterol transport: ABCA1 is a potential target for in utero therapy of Smith-Lemli-Opitz syndrome. Hum Mol Genet 2008; 17:3806-3813
  53. Sfefuji J, Panzenboeck U, Becker T, Hirschmugl B, Schweinzer C, Lang I, Marsche G, Sadjak A, Lang U, Desoye G, Wadsack C. Human endothelial cells of the placental barrier efficiently deliver cholesterol to the fetal circulation via ABCA1 and ABCG1. Circ Res 2009; 104:600-608
  54. Woollett LA. Maternal cholesterol in fetal development: transport of cholesterol from the maternal to the fetal circulation. Am J Clin Nutr 2005; 82:1155-1161
  55. Rosenson RS, Brewer HBJ, Davidson WS, Fayad ZA, Fuster V, Goldstein JL, Hellerstein M, Jiang X-C, Phillips MC, Rader DJ, Remaley AT, Rothblat GH, Tall AR, Yvan-Charvet L. Cholesterol efflux and atheroprotection:Advancing the concept of reverse cholesterol transport. Circulation 2012; 125:1905-1919
  56. Venkateswaran A, Laffitte BA, Joseph SB, Mak PA, Wilpitz DC, Edwards PA, Tontonoz P. Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXR alpha. Proc Natl Acad Sci 2000; 97:12097-12102
  57. Repa JJ, Turley SD, Lobaccaro JMA, Medina J, Li L, Lusting K, Shan B, Heyman RA, Dietschy JM, Manglesdorf DJ. Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 2000; 289:1524-1529
  58. Janowski BA, Grogan MJ, Jones SA, Wisely GB, Kliewer SA, Corey EJ, Mangelsdorf DJ. Structural requirements of ligands for the oxysterol liver X receptors LXRalpha and LXRbeta. Proc Natl Acad Sci 1999; 96:266-271
  59. Linton MF, Tao H, Linton EF, Yancey PG. SR-BI:A multifunctional receptor in cholesterol homeostasis and atherosclerosis. Trends Endocrinol Metab 2017; 28:P461-P472
  60. Leiva A, Verdejo HE, Benitez ML, Martinez A, Busso D, Rigotti A. Mechanisms regulating hepatic SR-BI expression and their impact on HDL metabolism. Atherosclerosis 2011; 217:299-307
  61. Jenkins KT, Merkens LS, Tubb MR, Myatt L, Davidson WS, Steiner RD, Woollett LA. Enhanced placental cholesterol efflux by fetal HDL in Smith-Lemli-Opitz syndrome. Mol Genet Metab 2008; 94:240-247
  62. Scholler M, Wadsack C, Metso J, Manavaian APC, Sreckovic I, Schweinzer C, Hiden U, Jauhiainen M, Desoye G, Panzenboeck U. Phospholipid transfer protein is differentially expressed in human arterial and venous placental endothelial cells and enhnaces cholesterol efflux to fetal HDL. J Clin Endocrinol Metab 2012; 97:2466-2474
  63. Scholler M, Wadsack C, Lang I, Etschmaier K, Schweinzer C, Marsche G, Dieber-Rotheneder M, Desoye G, Panzenboeck U. Phospholipid transfer protein in the placental endothelium is affected by gestational diabetes mellitus. J Clin Endocrinol Metab 2012; 97:437-445
  64. Melchior JT, Street SE, Andraski AB, Furtado JD, Sacks FM, Shute RL, Greve EI, Swertfeger DK, Li H, Shah AS, Lu LJ, Davidson WS. Apolipoprotein A-II alters the proteome of human lipoproteins and enhances cholesterol efflux from ABCA1. J Lipid Res 2017; 58:1374-1385
  65. Shah AS, Tan L, Long JL, Davidson WS. Proteomic diversity of high density lipoproteins: our emerging understanding of its importance in lipid transport and beyond. J Lipid Res 2013; 54:2575-2585
  66. Davidson WS, Cooke AL, Swerfeger DK, Shah AS. The difference between high density lipoprotein subfractions and subspecies:an evolving model in cardiovascular disease and diabetes. Curr Atheroscler Rep2021; 23:23
  67. Stadler JT, Wadsack C, marsche G. Fetal high-density lipoproteins:current knowledge on particle metabolism, composition and function in health and disease. Biomedicines 2021; 9:349
  68. Wolin E, White J, Pottala JV, Sasinowski M, Dall T, Dayspring TD, McConnell JP, Hoefner DM, Varvel SA, Thiselton DL, Warnick GR, Harris WS. Comparison of cardiometabolic risk biomarkers from a national clinical laboratory with the US adult population. J Clin Lipidol 2015; 9:817-823
  69. Merzouk H, Meghelli B, M., Loukidi B, Prost J, Belleville J. Impaired serum lipids and lipoproteins in fetal macrosomia related to maternal obesity. Biol Neonate 2000; 77:17-24
  70. Lindegaard MLS, Svarrer EMM, Damm P, Mathiesen ER, Nielsen LB. Increased LDL cholesterol and CRP in infants of mothers with type 1 diabetes. Diabetes Metab Res Rev 2008; 24:465-471
  71. Hou R-L, Jin W-Y, Chen X-Y, Jim Y, Wang X-M, Shao J, Zhao Z-Y. Cord blood C-peptide, insulin, HbA1c, and lipids levels in small- and large-for-gestational-age newborns. Med Sci Monitor 2014; 20:2997-2105
  72. Koklu E, Kurtoglu S, Akcakus M, Koklu S, Buyukkayhan D, Gumus H, Yikilmaz A. Increased aortic intima-media thickness is related to lipid profile in newborns with intrauterine growth restriction. Horm Res 2006; 65:269-275
  73. Nayak CD, Agarwal V, Nayad DM. Correlation of cord blood lipid heterogeneneity in neonates with their antropometry at birth. Ind J Clin Biochem 2013; 28:152-157
  74. Kelishadi R, Badiee Z, Adeli K. Cord blood lipid profile and associated factors baseline data of a birth cohort study. Ped Peri Epidem 2007; 21:513-526
  75. Nagano N, Okada T, Yonezawa R, Yoshikawa K, Fujita H, Usukura Y, Makimoto M, Hosono S, Fujita Y, Takahashi S, Mugishima H, Yamamoto T. Early postnatal changes of lipoprotein subclass profile in later preterm infants. Clin Chim Acta 2011; 413:109-112
  76. Gozlan O, Gross D, Gruener N. Lipoprotein levels in newborns and adolescents. Clin Biochem 1994; 27:305-306
  77. Legras B, Durou MR, Ruelland A, Galou G, Jezequel C, Cloarec L. Serum lipid, apolipoprotein and lipoparticle levels in the human fetus. Prenat Diagn 1995; 15:225=228
  78. Khoury J, Henriksen T, Christophersen B, Tomstad S. Effect of a cholesterol-lowering diet on maternal, cord, and neonatal lipids, and pregnancy outcomes: A randomized clinical trial. Am J Obstet Gynecol 2005; 193:1292-1301
  79. Sales WB, Dias SJJ, Kroll C, Mastroeni SSBS, Silva JC, Mastroeni MF. Influence of altered maternal lipid profile on the lipid profile of the newborn. Arch Endocrinol Metab 2015; 59:123-128
  80. Johnson HJJ, Simpson ER, Carr BR, NacDonald PC, Parker CRF. The levels of plasma cholesterol in the human fetus throughout gestation. Pediat Res 1982; 16:682-683
  81. Pecks U, brieger M, Schiessl B, Bauerschiang DO, Piroth D, Bruno B, Fitzner C, Orlikowsky T, Maass N, Rath W. Maternal and fetal cord blood lipids in intrauterine growth restriction. J Perinat Med 2012; 2012:287-296
  82. Shoji H, Murano Y, Mori M, Matsunaga N, Ohkawa N, Suganuma H, Ikeno M, Hisata K, Hirayama S, Ueno T, Miida T, Shimizu T. Lipid profile and atherogenic indices soon after birth in Japanese preterm infants. Acta Paediatr 2014; 103:22-26
  83. Parker CRJ, Carr BR, Simpson ER, MacDonald PC. Decline in the concentration of low-density lipoprotein-cholesterol in human fetal plasma near term. Metabolism 1983; 32:919-923
  84. Pecks U, Mohaupt MG, Hutten MC, Maass N, Rath W, Escher G. Cholesterol acceptor capacity is preserved by different mechanisms in preterm and term fetuses. Biochim Biophys Acta 2014; 1841:251-258
  85. Cai HJ, Xie CL, Chen Q, Chen XY, Chen YH. The relationship between hepatic low-density lipoprotein receptor activity and serum cholesterol level in the human fetus. Hepatology 1991; 13:852-857
  86. Ahn E-M, Cho S-C, Lee MG, Cha Y-S. Serum carnitine, triglyceride and cholesterol profiles in Korean neonates. Br J Nutr 2007; 98:373-379
  87. Gordon SM, Hofmann S, Askew DS, Davidson WS. High density lipoprotein: its not just about lipid transport anymore. Trends Endocrinol Metab 2011; 22:9-15
  88. Blum CB, Davis PA, Forte TM. Elevated levels of apolipoprotein E in the high density lipoproteins of human cord blood plasma. J Lipid Res 1985; 26:755-760
  89. Fujita H, Okada T, Inami I, Makimoto M, Hosono S, Minato M, Takahashi S, Mugshima H, Yamamoto T. Heterogeneity of high-density lipoprotein in cord blood and its postnatal change. Clin Chim Acta 2008; 389:93-97
  90. Sreckovic I, Bimer-Gruenberger R, Obrist B, Stojakovic T, Schamagl H, Holzer M, Scholler M, Philpose S, Marshe G, Lang U, Desoye G, Wadsack C. Distinct composition of human fetal HDL attenuates its anti-oxidative capacity. Biochim Biophys Acta 2013; 1831:737-746
  91. Gugliucci A, Numaguchi M, Caccavello R, Kimura S. Small-dense low-density lipoproteins are the predominant apoB-100-containing lipoproteins in cord blood. Clin Biochem 2014; 47:475-477
  92. Davidson WS, Shah AS. High-density lipoprotein subspecies in health and human disease:Focus on type 2 diabetes. Methodist Debkey Cardiovasc J 2019; 15:55-61
  93. Augsten M, Hackl H, Ebner B, Chemelli A, Glatter O, Marsche G, Lang U, Desoye G, Wadsack C. Fetal HDL/apoE: a novel regulator of gene expression in human placental endothelial cells. Physiol Genomics 2011; 43:1255-1262
  94. Beath SV. Hepatic function and physiology in the newborn. Sem Neonatology 2003; 8:337-346
  95. Balistreri W, Heubi J, Suchy F. Immaturity of the enterohepatic circulation in early life: factors predisposing to "physiologic" maldigestion and cholestasis. J Pediatr Gastro Nutr 1983; 2:346-354
  96. Turley SD, Burns DK, Rosenfeld CR, Dietschy JM. Brain does not utilize low density lipoprotein-cholesterol during fetal and neonatal development in the sheep. J Lipid Res 1996; 37:1953-1961
  97. Carr BR. Metabolism of lipoproteins by human fetal hepatocytes. Am J Obstet Gynecol 1987; 157:1338-1344
  98. Andersen GE, Johansen KB. LDL receptor studies in term and pre-term infants.Measurement of sterol synthesis in cord blood lymphocytes. Acta Pediatr Scand 1980; 69:577-580
  99. Carr BR, Simpson ER. Lipoprotein utilization and cholesterol synthesis by the human fetal adrenal gland. Endocr Dev 1981; 2:306j-326
  100. Porter FD. Human malformation syndromes due to inborn errors of cholesterol synthesis. Curr Opin Pediatr2003; 15:607-613
  101. Phillipi CA, Steiner RD. Genetics of disorders of cholesterol biosynthesis. 2008;
  102. Platt FM, Wassif CA, Colaco A, Dardis A, Lloyd-Evans E, Bembi B, Porter FD. Disorders of cholesterol metabolism and their unanticipated convergent mechanisms of disease. Annu Rev Genomics Hum Genet2014; 15:173-194
  103. Kanungo S, Soares N, He M, Steiner RD. Sterol metabolism disorders and neurodevelopment-an update. Dev Dis Res Rev 2013; 17:197-210
  104. Weed M, Mundlos S, Olsen BR. The role of sonic hedgehog in vertebrate development. Matrix Biol 1997; 16:53-58
  105. Marti E, Bovolenta P. Sonic hedgehog in CNS development: one signal, multiple outputs. Trends Neuro 2002; 25:89-96
  106. Cooper MK, Wassif CA, Krabowiak PA, Taipale J, Gong R, Kelley RI, Porter FD, Beachy PA. A defective response to Hedgehog signaling in disorders of cholesterol biosynthesis. Nature Genet 2003; 33:508-513
  107. Pecks U, Bornemann V, Klein A, Segger L, Maass N, Alkatout I, Eckmann-Scholz C, Elessawy M, Lutjohann D. Estimating fetal cholesterol synthesis rates by cord blood analysis in intrauterine growth restriction and normally grown fetuses. Lipids Health Dis 2019; 18:185
  108. Francis KR, Ton AN, Xin Y, O'Halloran PE, Wassif CA, Malik N, Willisma IM, Cluzeau CV, Trivedi NS, Pavan WJ, Cho W, Westphal H, Porter FD. Modeling Smith-Lemli-Opitz syndrome with induced pluripotent stem cells reveals a causal role for Wnt/β-catenin defects in neuronal cholesterol synthesis phenotypes. Nat Med 2016; 22:388-396
  109. Sikora DM, Pettit-Kekel K, Penfield J, Merkens LS, Steiner RD. The near universal presence of autism spectrum disorders in children with Smith-Lemli-Opitz syndrome. Am J Med Genet A 2006; 140:1511-1518
  110. Tierney E, Bukelis I, Thompson RE, Ahmed K, Aneja A, Kratz L, Kelley RI. Abnormalaties of cholesterol metabolism in autism spectrum disorders. Am J Med Genet B 2006; 141:666-668
  111. Luo YB, Eran A, Palmer N, Avillach P, Levy-Moonshine A, Szolovits P, Kohane IS. A multidimensional precision medicine approach identifies an autism subtype characterized by dyslipidemia. Nat Med 2020; 26:1375-1379
  112. Tierney E, Remaley AT, Thurm A, Jager LR, Wassif CA, Kratz L, Bailey-Wilson JE, Bukelis I, Sarphare G, Jung ES, Brand B, Noah KK, Porter FD. Sterol and lipid analyses identifies hypolipidemia and apolipoprotein disorders in autism associated with adaptive functioning deficits. Trans Psy 2021; 11:471
  113. Duhig K, Chappell LC, Shennan AH. Oxidative stress in pregnancy and reproduction. Obstet Med 2016; 9:113-116
  114. Chiarello DI, Abad C, Rojas D, Toledo F, Vazquez CM, Mate A, Sobrevia L, Marin RM. Oxidative stress:Normal pregnancy versus preeclampsia. Biochim BiophysActa - Mol Basis Dis 2020; 1866:165354
  115. Schieber M, Chandel NS. ROS Function in redox signaling and oxidative stress. Curr Biol 2014; 24:R453-R462
  116. Murphy RC, Johnson KM. Cholesterol, reactive oxygen species, and the formation of biologically active mediators. J Biol Chem 2008; 283:15521-15525
  117. Janowski BA, Willy PJ, Devi TR, Falch JR, Mangelsdorf DJ. An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 1996; 383:728-731
  118. Sever N, Mann RK, Xu LM, Snell WJ, Hernandez-Lara CI, Porter NA, Beachy PA. Endogenous B-ring oxysterols inhibit the Hedgehog component Smoothened in a manner distinct from cyclopamine or sid-chain oxysterols. Proc Natl Acad Sci 2016:Ahead of print
  119. Sun Y, Kopp S, Strutz J, Gali CC, Zandl-Lang M, Fanaee-Danesh E, Kirsch A, Cvitic S, Frank S, Saffery R, Bjorkhem I, Desoye G, Wadsack C, Panzenboeck U. Gestational diabetes mellitus modulates cholesterol homeostasis in human fetoplacental endothelium. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1863:968-979
  120. Mistry HD, Kurlak LO, Mansour YT, Zurkinden L, Mohaupt MG, Escher G. Increased maternal and fetal cholesterol efflux capacity and placental CYP27A1 expression in preeclampsia. J Lipid Res 2017; 58:1186-1195
  121. Catarino C, Rebelo I, Belo L, Rocha-Pereira P, Rocha S, Bayer Castro E, Patricio B, Quintanila A, Santos-Lilva A. Fetal lipoprotein changes in pre-eclampsia. Acta Obstet Gynecol 2008; 87:628-634
  122. Leon-Reyes RG, Sosa SE, Medina-Navarro R, Guzman-Grenfell AM, Medina-Urrutia AX, Fuentes-Garcia S, Hicks GJJ, Torres-Ramos YD. Oxidative modifications of foetal LDL-c and HDL-c lipoprotein in preeclampsia. Lipids Health Dis 2018; 17:110
  123. Kilby MD, Nary RH, Mackness MI, Durrington PN. Fetal and maternal lipoprotein metaolism in human pregnancy complicated by type I diabetes mellitus. J Clin Endocrinol Metab 1998; 83:1736-1741
  124. Aslsaweed M, Hartmann PE, Geddes DT, Kakulas F. MicroRNAs in breastmillk and the lactating breast:Potential immunoprotectors and developmental regulators for the infant and the mother. Int J Environ Res Public Health 2015; 12:13981-14020
  125. Mastromarino P, Capoblanco D, Campagna G, Laforgia N, Drimaco P, Dileone A, Baldsassarre ME. Correlation between lactoferrin and beneficial microbiota in breast milk and infant's feces. Biometals 2014; 27:1077-1086
  126. Carr LE, Viman MD, Rosa F, Munbit D, Matzel KS, Elolimy AA, Yenuva L. Role of human milk bioactives on infants' gut and immune health. Front Immunol 2021; 12:60400
  127. Babawale EA, Jones PJH, Mercer KE, Lin H, Yeruva L, Yoseph FB, Rutherfurd SM. Modulating sterol concentrations in infant formula influences cholesterol absorption and synthesis in the neonatal piglet. Nutrients 2018; 10:1848
  128. Friedman G, Goldberg SJ. Concurrent and subsequent serum cholesterols of breast- and formula-fed infants. Am J Clin Nutr 1975; 38:42-45
  129. Wagner V, Stockhausen HV. The effect of feeding human milk and adapted milk formulae on serum lipid and lipoprotein levels in young infants. Euro J Pediatr 1988; 147:292-295
  130. Demmers TA, Jones PJH, Wang Y, Krug S, Creutzinger V, Heubi JE. Effects of early cholesterol intake on cholesterol biosynthesis and plasma lipids among infants until 18 months of age. Pediatrics 2005; 115:1594-1601
  131. Bayley TM, Alasmi M, Thorkelson T, Krug-Wispe S, Hones PJH, Bulani JL, Tsang RC. Influence of formula versus breast milk on cholesterol synthesis rates in four-month-old infants. Pediatr Res 1998; 44:60-67
  132. Yao L, Woollett LA. Adult sterol metabolism is not affected by a positive sterol balance in the neonatal Golden Syrian hamster. Am J Physiol 2005; 288:R561-R566
  133. Barker DJP, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. The Lancet1986; 1(8489):1077-1081
  134. Barker DJP. The intra-uterine origins of disturbed cholesterol homeostasis. Acta Paediatr 1999; 88:483-492
  135. Thornburg KL, Louey S. Fetal roots of cardiac disease. Heart 2005; 91:867-868
  136. thousanddays.com.
  137. Yang F, Janszky I, Gisslr MR, N., Wikstrom A-K, Yu Y, Chen H, Edstedt Bonamy A-K, Li J, Laszlo KD. Association of maternal preeclampsia with offspring risks of ischemic heart disease and stroke in Nordic countries. JAMA Netw Open 2022; 5:42064
  138. Lu HQ, Hu R. Lasting effects of intrauterine exposre to preeclampsia on offspring and the underlying mechanism. Am J Perinat Rep 2019; 9:e275-e291
  139. Frost AL, Suriano K, Aye CYL, Leeson P, Lewandowski AJ. The immediate and long-term impact of preeclampsia on offspring vascular and cardiac physiology in the preterm infant. Front Pediatr 2021; 9:625726
  140. Wojczakowski W, Kimber-Trojnar Z, Dziwisz F, Slodzinska M, Slodzinski H, Leszczynska-Gorzelak B. Preeclampsia and cardiovascular risk for offspring. J Clin Med 2021; 10:3154
  141. Lewandowski AJ, Levy PT, Bates ML, McNamara PJ, Nuyt AM, Goss KN. Impact of the vulnerable preterm heart and circulation on adult cardiovascular disease risk. Hypertension 2020; 76:1028-1037
  142. Crump C, Howell EA, Stroutstrup A, McLaughlin MA, Sundquist J, Sundquist K. Association of preterm birth with risk of ischemic heart disease in adulthood. JAMA Pediatr 2019; 173:736-743
  143. Wu P-H, Gulati M, Kwik CS, Wong CW, Narain A, O-Brien S, Chew-Graham CA, Verma G, Kadam UT, Mamas MA. Preterm delivery and future risk of maternal cardiovascular disease: a systematic review and meta-analysis. J Am Heart Assoc 2018; 7:e007809
  144. Zhang X, Ho SM. Epigenetics meets endocrinology. J Mol Endocrinol 2011; 46:R11-R32
  145. Linner A, Almgren M. Epigenetic programming-The important first 1000 days. Acta Paediatr 2020; 109:443-452
  146. Schiano C, Vieri MT, Grimaldi V, Pisacia A, Pascale MR, Napoli C. Epigenetic-related therapeutic challenges in cardiovascular disease. Trends Pharmacol Sci 2015; 36:226-235
  147. Sen S, Simmons RA. Maternal antioxidant supplementation prevents adiposity in the offspring of Western diet-fed rats. Diabetes 2010; 59:3058-3065
  148. Agostoni C, Baselli L, Mazzoni MB. Early nutrition patterns and diseases of adulthood: a plausible link? Eur J Intern Med 2013; 24:5-10
  149. Marmot MG, Page CM, Atkins E, Douglas JWB. Effect of breast-feeding on plasma cholesterol and weight in young adults. J Epidemiol Community Health 1980; 34:164-167
  150. Parikh N, Hwang SJ, Ingelsson E, Benjamin EJ, Fox CS, Vasan RS, Murabito JM. Breastfeeding in infancy and adult cardiovascular disease risk factors. Am J Med 2009; 122:656-663
  151. Kolacek S, Kapetanovic T, Zimolo A, Luzar V. Early determinants of cardiovascular risk factors in adults. A. Plasma lipids. Acta Paediatr 1993; 82:699-704
  152. Hodgson PA, Ellefson RD, Elvebeck LR, Harris LE, Nelson RA, Weldman WH. Comparison of serum cholesterol in children fed high, moderate, or low cholesterol milk diets during neonatal period. Metabolism1979; 25:739-746
  153. Owen CG, Whincup PH, Kaye SJ, Martin RM, Davey Smith G, Cook DG, Bergstrom E, Black S, Wadsworth ME, Fall CHD, Freudenheim J, Nie J, Huxley RR, Kolacek S, Leeson CP, Pearce MS, Raitakari OT, Lisnen I, Vilkari JS, Ravelli AC, Rudnicka AR, Strachan DP, Williams SM. Does initial breastfeeding lead to lower blood cholesterol in adult life? A quantitative review of the evidence. Am J Clin Njutr 2008; 88:305=314
  154. Colen CG, Ramey DM. I breast truly best?Estimating the effect of breastfeeding on long-term child wellbeing in the United States using sibling comparisons. Soc Sci Med 2014; 109:55-65
  155. Vuorio AF, Turtola H, Kontula K. Neonatal diagnosis of familial hypercholesterolemia in newborns born to a heterozygous familial hypercholesterolemia. Arterioscler Thromb Vasc Biol 1997; 17:3332-3337
  156. Pathirana MM, Lassi ZS, Roberts CT, Andraweera PH. Cardiovascular risk factors in offspring exposed to gestational diabetes mellitus in utero:systematic review and meta-analysis. J  Dev Orig Health Dis 2020; 11:599-616
  157. Higa R, Leonardi ML, Jawerbaum A. Intrauterine programming of cardiovascular diseases in maternal diabetes. Front Physiol 2021; 12:760251
  158. Cooper R, Pinto Pereira SM, Power C, Hypponen E. Parental obesity and risk factors for cardiovascular disease among their offspring in mid-life: findings from the 1958 British Birth Cohort Study. Int J Obes 2013; 37:1590-1596
  159. Razaz N, Villamor E, Muraca GM, Edstedt Bonamy A-K, Cnattingius S. Maternal obesity and risk of cardiovascular diseases in offspring: a population-based cohort and sibling-controlled study. Lancet Diabetes Endocrinol 2020; 8:572-581

 

Fine-Needle Aspiration of the Thyroid Gland

ABSTRACT

 

Thyroid nodules are common in clinical practice and the majority are benign with the risk of malignancy varying from 7 to 15%. Clinical evaluation includes careful history and physical examination, laboratory tests, neck ultrasound (US), and a fine-needle aspiration (FNA). Thyroid FNA or biopsy is an accurate test for determining malignancy in a nodule and is an integral part of current thyroid nodule evaluation. Results are superior when FNA is performed with ultrasound-guidance (USFNA). Herein, we describe techniques used for US-guided FNA. FNA results are classified as diagnostic (satisfactory) or nondiagnostic (unsatisfactory). Unsatisfactory smears (5-10%) result from hypocellular specimens usually caused by cystic fluid, bloody smears, or suboptimal preparation. Diagnostic smears are conventionally classified into benign, indeterminate, or malignant. A benign cytology is negative for malignancy, and includes cysts, colloid nodule, or Hashimoto thyroiditis. Malignant (or suspicious for malignancy) cytology is usually positive for malignancy on histology, and includes primary thyroid tumors or, less frequently, nonthyroidal metastatic cancers. Papillary thyroid carcinoma (PTC) is the most common malignancy, characterized by increased cellularity, sheets of cells, and typical nuclear abnormalities. Indeterminate or suspicious specimens include atypical changes, Hürthle (oncocytic) cells or follicular neoplasms, typically with absent or scant colloid, hypercellularity, and sometimes a microfollicular arrangement. The Bethesda Cytologic Classification has a 6-category classification. Overall, the indeterminate category Bethesda III category has a risk of malignancy of 6-18% if Non-invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP) is not considered cancer. Advances in molecular testing can help further separate benign from malignant nodules with an indeterminate cytology.

 

INTRODUCTION

 

Thyroid nodules are common in clinical practice with a prevalence of up to 60%. The majority is benign and the risk of malignancy is between 7 to 15% (1).

 

Fine-needle aspiration biopsy (FNAB) of the thyroid gland is an accurate diagnostic test used routinely in the initial evaluation of nodular thyroid disease (2-6). Epidemiologic studies suggest that nodular thyroid disease is a common clinical problem, with a prevalence of 4% to 7% in the adult population in North America and an annual incidence of 0.1%, which translates into approximately 300,000 new nodules in the United States (3). In patients with a single palpable nodule, additional nodules can be detected in about 20-48% by ultrasonography (7).

 

A survey of clinical members of the American Thyroid Association revealed that most endocrinologists (96%) perform FNA for diagnosis of thyroid nodules (8). In addition, FNA with ultrasonographic guidance (US-FNA) is used routinely in follow-up surveillance of patients with thyroid cancer. Therefore, the importance of FNA biopsy in thyroid practice cannot be overemphasized.

 

This chapter describes biopsy techniques, cytologic diagnosis, complications, FNA results, diagnostic pitfalls, and other information that may be useful to clinicians who manage patients with nodular thyroid disease.

 

DEFINITIONS/HISTORY

 

The diagnosis of thyroid nodules by needle biopsy was first described by Martin and Ellis (9) in 1930, who used an 18-gauge–needle aspiration technique. Subsequently, cutting needle biopsy with Silverman or Tru-Cut needles was used for tissue examination. None of these techniques gained wide acceptance because of fear of malignant implants in the needle track, false-negative results, and serious complications. However, Scandinavian investigators introduced small‑needle aspiration biopsy of the thyroid in the 1960s, and this technique came into widespread use in North America in the 1980s (10).

 

For FNA biopsy, most use fine or thin (22- to 27-gauge) needles, most commonly 25 or 27-gauge needles. As the name indicates, the biopsy technique uses aspiration to obtain cells or fluid from a mass. In contrast to percutaneous large‑needle biopsy, which obtains tissue specimens and requires histologic fixation, aspiration biopsy offers cytological examination of the specimen. Another technique, fine‑needle non-aspiration (FNNA) biopsy, also referred to as capillary technique, avoids aspiration but obtains representative cytologic samples.

 

Although the FNA technique appears simple, considerable time and experience are required to acquire and maintain skillful biopsy technique. Debate continues about who is best qualified to perform FNA biopsy, but the best results are obtained if the person performing the biopsy has appropriate technique and volume. In the opinion of the authors, individuals performing biopsies should have appropriate knowledge of thyroid pathologies in order to relate the findings to the clinical context.

 

EQUIPMENT

 

The basic equipment needed to perform FNA biopsy is simple and relatively inexpensive (2, 4, 11) The following items are necessary to have but many are not essential (Table 1).

 

Table 1. FNA Biopsy Equipment – Figure 1

1.

Disposable 3-10-mL plastic syringes

2.

Disposable 25- or 27-gauge needles, 1.5 inches long [shorter needles can be used for more superficial nodules]

3.

Alcohol prep sponges

4.

Sterile gauze

5.

Gloves

6.

Probe cover

7.

Sterile gel

8.

Lidocaine—1% for those who prefer biopsy with local anesthesia

9.

Glass slides, with 1 end frosted on 1 side, 1-mm thin (Gold Seal, Erie Scientific Company)

10.

Alcohol swabs and bandaid for post procedure

 

Additional tools: For sample procesing

11.

Containers for (cystic) fluid collection and transportation to the cytology laboratory

12.

Laboratory slips with the patient’s name, clinic number, biopsy sites, and other relevant information to be transferred to the cytology laboratory

13.

Alcohol bottles for immediate wet fixation of smears

14.

Rarely used nowadays: A syringe holder or syringe pistol—most commonly used is the Cameco syringe pistol (Belpro Medical). The pencil-grip syringe holder is another syringe-holding device (Tao and Tao Technology, Incorporated).-

 

Figure 1. FNA biopsy equipment is simple and inexpensive. It includes alcohol wipes or disinfectant, gauze pads, plastic syringes, 23- to 27-gauge needles, glass slides, a fixative solution, and, optionally a pistol- grip mechanical syringe holder. Many experts use cell-preserving solutions to wash out the needles for the preparation of cytoblocs.

THE PATIENT

 

The thyroid gland should be palpated carefully and the nodule(s) to be biopsied identified under sonographic guidance. The consent process is important. It should fully explain the procedure including risks and benefits, and address all questions raised by the patient. The procedure can be done with local anesthesia or without. Per nodule, typically 3 informative passes are obtained, in particular if rapid on-site evaluation (ROSE) by a cytopathologist is not available.  In experienced hands, no serious complications are expected. Mild pain, minor bleeding or infection can occur but are unlikely to happen. Very rarely, transient hoarseness due to recurrent laryngeal nerve injury has been reported. With the appropriate caution, the procedure can usually be performed in patients with blood thinners.

 

The procedure is performed in the outpatient setting with the patient lying on an adjustable examination bed or chair. The procedure can be performed by a single person, but the presence of a nurse or clinical assistant is encouraged, if available. The patient may be seated or supine; we prefer the supine position. The patient is placed supine with the neck hyperextended to expose the thyroid; for support, a pillow is placed under the shoulders (Fig. 2 A). The patient is asked not to swallow, talk, or move during the procedure. It is best to talk to the patient and keep them informed of the progress of the biopsy. Once completed, firm pressure is maintained on the biopsy site(s) then the area is cleaned and a Band-Aid applied. It is best to observe patients for a few minutes for any side effects. This time can be used to counsel on after-procedure care instructions and potential results, if available including written instructions. Subsequently, the patient can be discharged.

Figure 2. A) Patient position during fine-needle aspiration (FNA). A supine position and a pillow under the patient’s shoulder allow hyperextension of the neck and maximal exposure. B) Syringe is placed in syringe holder. C) The nodule is identified and stabilized with operator’s the nonaspirating hand. The operator stands on the side of the patient opposite that of the thyroid nodule. Current Occupational Safety and Health Administration regulations require the use of gloves because of concern about bloodborne diseases. D) With a quick motion, the needle passes through the skin and enters the nodule. Immediate mild suction follows. As soon as aspirate appears, suction is released and the needle is withdrawn.

THE TECHNIQUES

 

FNA Biopsy

 

Numerous reports, reviews, and even textbooks provide detailed descriptions of various FNA biopsy techniques (11-15). Although most reports agree on the principles of the technique, variations have been described to improve results. It is important to position the patient correctly, identify and locate the mass, provide adequate light during the biopsy, and have a clinical assistant available for help if needed. The physician performing the biopsy should be positioned at the patient’s side, preferably contralateral to the lesion. The nodule(s) to be aspirated is identified, and the overlying skin is cleansed with alcohol. The use of povidone-iodine or sterile technique is not necessary but encouraged.

 

A retrospective study across multiple sites of the Mayo Clinic compared ultrasound-guided thyroid FNA using no anesthetic, subcutaneous injectable anesthetic, and topical anesthetic to compare the degree of pain/discomfort (16). The study found patient discomfort associated with FNA was comparable during and after the procedure regardless of the use of anesthetic or the type. However, others reported that FNA-associated pain is frequent and that the use of local anesthesia is beneficial (17)

 

In the method illustrated here, a 10‑mL plastic syringe is attached to a Cameco syringe holder and held in the right hand by a right‑handed operator (Fig. 2 B). Two fingers of the free (left) hand firmly grasp the nodule while the other hand holds a pistol-grip syringe holder (Fig. 2 C). The needle is then inserted through the skin and into the nodule. Once the needle tip is in the nodule, gentle suction is applied while the needle is moved in and out within the nodule vertically (Fig. 2 D). This maneuver allows the dislodging of cellular material and easy suction into the needle. During this period of 5 to 10 seconds, suction is maintained, and as soon as fluid or aspirate appears in the hub of the needle, the suction is released, and the needle is withdrawn. Of note, many operators do not use the handle; in this case, simply using a syringe with needle may suffice using the same maneuver.

 

Nowadays, ultrasound evaluation identifies cystic and partially cystic lesions. The appearance of fluid suggests that the nodule is cystic; suction is maintained, and as much fluid as possible is aspirated. It is important to release the syringe plunger and remove the vacuum before withdrawing the needle; this allows the aspirate to remain in the needle and not be sucked into the syringe. Next, the needle is detached from the syringe (Fig. 3 A), and 5 mL of air is drawn into the syringe (Fig. 3 B). The needle is reattached to the syringe, and with the bevel facing down, 1 drop of aspirated material is ejected onto each of several glass slides (Fig. 3 C). It is important that all slides be labeled and placed in order on a nearby table before the aspiration begins.

 

Smears are prepared by using a second glass slide in a manner similar to that of making blood smears (Fig. 3 D). The slides for wet fixation should be placed immediately in 95% alcohol for staining with the Papanicolaou stain. For Giemsa staining, air-dried smears are necessary, and prepared slides are left unfixed and transported to the laboratory.

 

Rapid on-site evaluation (ROSE) of cytology slides to evaluate for adequacy by cytopathologists may lower non-diagnostic rates and the number of necessary needle passes (18). This may, however, add more time to complete the procedure.

 

Figure 3. A) The needle is removed quickly from the syringe. B) Five ml of air is aspirated into the syringe, and the needle is placed back on the syringe. C) With the needle bevel facing down, 1 drop of aspirated material is expelled onto each of several glass slides. Slides are labeled and placed on the table before aspiration, ready for use. D) With a second slide, smears are prepared in a manner similar to that for blood smears. Slides are then immediately wet fixed by placing them in an alcohol bottle.

Usually, 3 to 6 aspirations are made (13, 14), some authors suggest at least 6 (19). Additional passes might be needed for molecular testing if decisions were made to obtain those. However, needle washouts typically provide sufficient materials for molecular analyses.  Preferably, the aspirates should be obtained from the peripheral areas and different parts of the nodule in a sequential manner to ensure representative sampling (13). For larger nodules, the deep center of the mass should be avoided because it is more likely to contain degeneration and fluid, decreasing the chance of a diagnostic specimen. For cystic lesions, the fluid should be completely aspirated and FNA attempted on residual solid tissue if present. Aspirated fluid should be placed in a plastic cup and saved for cytology evaluation. For each pass, one should use a new needle and syringe.

 

The needle can be inserted in parallel or perpendicular way to the ultrasound probe. In the parallel approach, the US image can see the entire needle path. The perpendicular approach is generally simpler and the needle travels shorter distance; however, only needle tip can be seen in the nodule. Both techniques are equally effective.

 

Generally, minor procedures such as thyroid FNA, can be safely performed while patients are on antiplatelets/anticoagulation (20). Special attention should be made to use a smaller gauge needle (27-gauge needle), minimize the number of passes, and monitor the patient for a few minutes for signs of bleeding. In certain situations, INR might need to be less than 2- 2.5 at the day of the procedure per certain operator preferences. Overall, the risk of stopping anticoagulation for the purpose of FNA procedure must always be weighed against the risk of complication related to discontinuing the medication for few days. The overall data suggest low risk of bleeding when doing the biopsy on anticoagulation and antiplatelet therapy with hematoma only occurred in 0.89% (21).

 

FNNA Biopsy

 

The FNNA, also referred to as fine-needle capillary (FNC) sampling technique, has been described by several authors (2, 22). This technique is thought to minimize trauma to thyroid tissue and to reduce blood contamination. For this technique, patient preparation is similar to that for FNA. However, no syringe or suction is necessary. The hub of a 27-gauge needle is held in a pencil-grip fashion, and the needle is gently inserted into the nodule and then moved in and out for 5 to 10 seconds. Aspirate flows into the needle through capillary action, and as soon as aspirate appears in the hub, the needle is withdrawn and attached to a syringe with air inside. Next, the plunger is used to expel the material onto glass slides. The procedure is repeated several times, and the slides are prepared as described above for FNA.

 

Other techniques such as Core-needle biopsy (23) or Large-needle aspiration biopsy (24) are used less commonly as first-line procedure. They can be associated with a higher risk of complications such as pain and bleeding, but they have some utility with repeatedly non-diagnostic FNA or when lymphoma, which requires additional analyses such as flow cytometry, is suspected.

 

POST BIOPSY PROCEDURE

 

After the biopsy has been completed, firm pressure is applied to the biopsy site(s) with a 4×4-inch gauze pad. Once bleeding has stopped, an adhesive bandage is placed on the puncture site(s), and the patient is observed for a few minutes. If there are no problems, the patient is allowed to leave.

 

COMPLICATIONS

 

Thyroid FNA biopsy, particularly using US-FNA, is very safe. No serious complications such as tumor seeding, nerve damage, tissue trauma, or vascular injury have been reported (11-15). Needle puncture may cause slight discomfort and possible skin ecchymosis at the aspiration site(s). However, even a minor hematoma is uncommon. Patient use of anticoagulants or salicylates does not preclude FNA biopsy. Needle track implantation of thyroid carcinoma is extremely rare and appears to be an exceptional complication (25). Post aspiration hemorrhage within a cystic lesion is uncommon.

 

CYTOLOGIC DIAGNOSIS

 

Aspirates from normal glands often have scant thyroid follicular cells and colloid. Wet‑fixed smears are usually prepared with a modified Papanicolaou stain, which shows nuclear details. Air-dried smears are often prepared with a Romanovsky stain. May‑Grünwald-Giemsa is a modified Romanovsky staining procedure that is sometimes used in thyroid cytologic preparations. The six Bethesda Classification System categories of cytology results include: Non-diagnostic (unsatisfactory), Benign, Atypia or undetermined significance (or follicular lesion of undetermined significance), Follicular neoplasm or suspicious for follicular neoplasm, suspicious for malignancy, and malignant (Table 7).

 

Benign Cytology

 

Aspirates obtained from multinodular goiters, a benign microfollicular adenoma, or normal thyroid are referred to as colloid nodules and show loosely cohesive sheaths of follicular epithelium, colloid, blood, and rare macrophages. Colloid nodules are the most common cytology and contain an abundance of colloid with sparse follicular cells. There is considerable variation in the number of cells, as well as the type and amount of colloid present (Fig. 4).

Figure 4. Colloid nodule. Sheath of normal thyroid epithelium shows uniform nuclei and pale cytoplasm (Papanicolaou, ×100).

Another benign diagnosis is Hashimoto thyroiditis. It has typically a characteristic pattern on FNA smears, showing hypercellularity with lymphocytes, Hürthle cells, and minimal or no colloid (Fig. 5).

Figure 5. Hashimoto thyroiditis. A) Group of Hürthle cells with large cytoplasm and prominent nuclei, surrounded by a teratogeneous population of lymphocytes (Papanicolaou, ×60). B) Hypercellular aspirate with lymphocytes and Hürthle cells (May-Grünwald-Giemsa, ×250).

Subacute (granulomatous) thyroiditis is a rare condition with a benign aspirate. Typically, the smear shows multinucleated giant cells, epithelioid histiocytes, and scattered inflammatory cells.

 

Malignant Cytology

 

Papillary carcinoma, the most common thyroid malignancy, is readily diagnosed by FNA. Typically, cytology shows large irregular nuclei, and nuclear grooves. Psammoma bodies may or may not be present, but if present, they are highly suggestive of papillary thyroid carcinoma (Fig. 6).

Figure 6. Papillary thyroid carcinoma. A) Follicular cells with large irregular nuclei, nuclear grooving, and pale chromatin (Papanicolaou, ×400). B) Histologic preparation showing typical papillary configurations (hematoxylin-eosin, ×50).

Medullary thyroid carcinoma accounts for 5% to 10% of thyroid cancers and may present as a thyroid nodule or neck mass. Typically, aspirates from a medullary thyroid carcinoma are hypercellular, composed of large, poorly cohesive cells, and predominantly spindle shaped. Amyloid is often, but not invariably, present, and there is no colloid (Fig. 7).

Figure 7. Medullary thyroid carcinoma. A) Cellular specimen staining positively for calcitonin by immunohistochemistry (×100). B) Loosely cohesive fragments of spindle-shaped cells. Amyloid is present as amorphous blue material intimately associated with neoplastic cells (Papanicolaou, ×400).

High-grade carcinoma can be diagnosed cytologically, but distinguishing between primary and metastatic cancer is not easy.

 

FNA RESULTS

 

The accumulated experience of the past 4 decades has confirmed the reliability and usefulness of FNA as a diagnostic test (2-4, 11-15, 26-31). The role of FNA biopsy in the evaluation of thyroid nodules is now firmly established, and FNA has become the initial test because it is both safe and cost effective. In most clinics, FNA has become a standard test, performed most often by an endocrinologist.

 

Diagnostic Cytology

 

An adequate specimen of good technical quality is considered diagnostic or satisfactory and may be benign, suspicious, or malignant. A benign (negative) cytologic diagnosis is reported for 50% to 90% of the specimens (average, 70%) (13, 15, 27, 32), 10-30% of FNA cytologic specimens may be suspicious for malignancy (indeterminate) (average, 20%) (32, 33). A malignant (positive) cytologic diagnosis is made in about 1% to 10% of cases (average, 5%). For example, Caruso and Mazzaferri (33) reported the following results from 9 series that included more than 9,000 patients: benign, 74%; malignant, 4%; inadequate, 11%; and suspicious, 11%. We reviewed more than 18,000 specimens from 7 large series and obtained similar cytologic results: benign, 69%; malignant, 4%; suspicious, 10%; and nondiagnostic, 17% (32).

 

False Negative Rates

 

False-negative results reflects missed malignancy. False-negative rates generally occur in 1.5% to 11.5% (average, <5%) (19, 30, 33, 34). The false-negative rate is defined as the percentage of patients with a benign cytology in whom malignant lesions are later confirmed on thyroidectomy. The frequency of false-negative cytologic diagnosis depends on the number of patients who subsequently have surgery and histologic review. In most retrospective series, less than 10% of patients with a benign cytologic diagnosis subsequently undergo thyroid surgery, suggesting that false-negative rates should be interpreted with some skepticism (33, 35). Despite this note of caution, most authorities agree that the true false-negative rate is less than 5% if all patients undergo thyroid surgery. False-negative rates are lower in centers experienced with the procedure and with cytologic interpretation by expert cytopathologists.

 

False Positive Rates

 

False-positive rates vary from 0% to 8% (average, 3%) (30, 32, 33). A false‑positive diagnosis indicates that a patient with a malignant FNA result was found on histologic examination to have benign lesions.

 

Causes of False Diagnoses

 

Interpretive or sampling errors account for false diagnoses (14, 32, 34). Hashimoto thyroiditis is probably the most common cause of a false‑positive cytology. Misclassification of follicular and Hürthle cell adenomas as papillary carcinomas accounts for other errors. FNA biopsy of thyroid lymphomas may yield lymphocytes that can be interpreted as Hashimoto thyroiditis, accounting for a false‑negative diagnosis. Inadequate or improper sampling accounts for some false-negative errors. For example, nodules smaller than 1 cm may be too small for accurate needle placement, and nodules larger than 4 cm may be too large to allow proper sampling from all areas, thereby increasing the likelihood of misdiagnosis. Finally, the cytopathologist should establish and observe criteria to exclude a diagnosis of malignancy (2, 4, 11, 26).

 

The Problem of Cellular Tumors

 

Hypercellular specimens from follicular or Hürthle cell lesions may have features suggestive of, but not diagnostic for malignancy (3, 11, 14, 27). Thus, the cytopathologist labels these suspicious for malignancy because cytologic features neither confirm nor rule out malignancy. Histologic examination is necessary for definitive diagnosis. Hypercellularity may be seen with non-neoplastic lesions, and Hürthle cell changes may be seen in patients with lymphocytic thyroiditis. The diagnosis of follicular neoplasm is indicative of an underlying malignancy in 14% of cases and Hürthle cell neoplasm in 15% (29, 32). Many pathologists maintain that benign and malignant follicular tumors cannot be distinguished on the basis of aspirated cells only, and the lesion must be removed for histopathologic examination (4, 14, 35). However, Kini et al (36) believes that follicular adenomas and follicular carcinomas usually can be differentiated on the basis of nuclear size.

 

Several authors have discussed the problem of follicular neoplasm. In a study of 149 patients with the cytologic diagnosis of follicular neoplasm, Tuttle et al. (37) reported that risk of malignancy was higher in men, solitary nodules, and nodules larger than 4 cm. In a study of 219 patients with follicular neoplasm, Schlinkert et al. (38) showed that nodules are more likely malignant in younger patients, in men, if the nodule is solitary, and if it is larger than 4 cm. Baloch et al. (39) studied 184 cases of follicular neoplasm and reported that risk factors for malignancy included male sex, older age (>40 years), and larger nodules (>3 cm). Overall, they found that 70% of these lesions are benign.

 

Molecular studies are now routinely used with thyroid nodule evaluation and biopsy to help further asses the likelihood of malignancy in thyroid nodules in nodules with undetermined cytology with reasonable sensitivity and specificity [see section below].

 

Non-Diagnostic Cytology

 

Inadequate specimens are labeled non-diagnostic or unsatisfactory and account for 2% to 20% of specimens (average, 10%) (32, 33). Several factors influence non-diagnostic rates for FNA results, including the skill of the operator, vascularity of the nodule, criteria used to judge adequacy of the specimen, and the cystic component of the nodule (40-43). Overall, a satisfactory smear contains at least 6 clusters of well‑preserved cells, with each group consisting of at least 10 to 15 cells. Re-aspirations with US-FNA yields satisfactory specimens in more than 50% of cases with a non-diagnostic initial FNA (19, 35).

 

Chow et al. found a 7% malignancy rate in 153 patients with initial non-diagnostic smears (44). Among 27 patients treated surgically, 37% had cancer. Re-aspiration with US-FNA was diagnostic in 66% and 56% without US; overall, 62% of re-aspirations were diagnostic.

For patients with non-diagnostic FNA biopsy, FNA can be repeated few weeks later. A repeat ultrasound-guided FNA will yield a diagnostic cytology specimen in 75% of solid nodules and 50% of cystic nodules (45).

 

If a patient has two nondiagnostic FNAs, ultrasound-guided core-needle biopsy can be considered as it has higher diagnostic yield compared to further repeating FNA (46, 47). However, the prevalence of cancer in non-diagnostic results is lower than in the general population. Nodules with repeatedly non-diagnostic FNA results typically have a low risk of malignancy and can be monitored if suspicious sonographic features are lacking (48).

 

DIAGNOSTIC ACCURACY

 

Analysis of the data reveals that the sensitivity of FNA ranges from 65% to 98% (mean 83%), and specificity ranges from 72% to 100% (mean 92%) (13, 30, 33). The predictive value of a positive or suspicious cytologic result is approximately 50%. The overall accuracy for cytologic diagnosis approaches 95% (Table 2).

 

 

Table 2. Summary Data From Literature Survey on Thyroid FNA

Feature

Mean

Range

Definition

Sensitivity (%)

83

65-98

Likelihood that a patient who has disease has a positive test result

Specificity (%)

92

72-100

Likelihood that a patient without disease has a negative test result

Positive predictive value (%)

75

50-96

Fraction of patients who have a positive test who have disease

False-negative rate (%)

5

1-11

FNA negative; histology positive for cancer

FNA, fine-needle aspiration.

*From Gharib et al. (4). Used with permission.

 

 

FNA GUIDELINES

 

Several guidelines and reviews have been published to help improve the adequacy and accuracy of cytology specimens (4, 49-51).

 

FNA biopsy should be performed by individuals who have had training in both thyroid ultrasound examination and thyroid biopsy. Thyroid FNA in the hands of experienced operators achieves high diagnostic accuracy. Aspirates should be obtained from different portions of the nodule, preferably peripheral areas, in an organized and sequential manner. It is essential to ensure that an adequate number of follicular cells are present. A cytopathologist, preferably one with experience in thyroid cytology, should review and interpret the slides. If re-aspiration yields insufficient material, a core biopsy is the next step. In the event that the final result is still insufficient, surgical excision is warranted for nodules with suspicious features.

 

Several reports have offered suggestions to minimize false-negative rates (3, 4, 19, 32). In a review on thyroid FNA, Belfiore and La Rosa suggested acquiring biopsy expertise, avoid suboptimal sampling, and repeat FNA during follow up especially if sonographically suspicious to reduce false-negative results (11).

 

To minimize false‑negative results, we follow the steps summarized in Table 3.

 

Table 3. Steps to Improve Accuracy of FNA and Lead to Better Nodule Management

Step

Explanation

Biopsy performed by expert in thyroid pathologies

Offers better thyroid examination; accumulates experience with FNA

Experienced cytopathologist reviews slides

Improves cytologic interpretation

Caution with small (<1 cm) or large (>4 cm) nodules

Increased chance of misdiagnosis;

US FNA improves accuracy

3-6 aspirates from different nodule sites

Improves cytologic sampling

Re-biopsy if cytology is nondiagnostic

About 50% will be diagnostic on re-aspiration

Nondiagnostic cytology is not negative

Risk of cancer is low but not ruled out

Aspirates with no follicular cells are nondiagnostic

These should not be considered negative for malignancy

Excise nodules yielding suspicious cytology

20-40% chance of malignancy

Excise clinically suspicious, cytologically benign nodules

Clinical impression overrides FNA diagnosis

FNA, fine-needle aspiration; US-FNA, FNA with ultrasonographic guidance.

*Modified from Gharib (2). By permission of Mayo Foundation for Medical Education and Research.

 

US-FNA BIOPSY

 

Published thyroid guidelines and reviews state that thyroid US should not be used as a screening test in the general population (3, 4). However, US is recommended for all patients with a single palpable nodule or a multinodular goiter or in a patient suspected of having a nodule (3) (Table 4). US machines are safe, easy to use, relatively inexpensive, have high resolution, and are widely available. It is important to note that US results are quite operator dependent.

 

Table 4. Indications for Thyroid Ultrasound Examination*

Palpable solitary nodule

Palpable multinodular goiter

Suspicion of nodule(s) in patient with difficult neck palpation

Prior history of neck radiation

Family history of medullary thyroid carcinoma, multiple endocrine neoplasia type 2, or papillary thyroid carcinoma

Unexplained cervical adenopathy

Preoperative thyroidectomy for cancer; long-term postoperative surveillance

*Data from Gharib et al. (4).

 

THYROID ULTRASOUND AND SONOGRAPHIC RISK STRATIFICATION SYSTEMS

 

Thyroid US is the imaging modality of choice for the evaluation of thyroid nodule(s). High-resolution neck US is safe and provides excellent imaging results with no significant cost, and can be done in the outpatient settings. Sonographic features used to characterize thyroid nodule risk of malignancy focus on five major sonographic criteria: echogenicity, composition, shape, echogenic foci, and margins (Table 5). Vascularity is debatable and therefor, not currently included in those criteria. US can also identify concerning lymph nodes. Features such as hypoechogenicity, irregular/infiltrative border, microcalcification, and a taller-than-wide shape in the transverse view, individually or in combination, are suggestive of malignancy especially if cervical lymph node involvement is seen. The diagnostic sensitivity and specificity of each of these features is variable and none of them alone can reliably distinguish malignant lesion from otherwise (52).

 

Table 5. Sonographic Characteristics of Thyroid Nodules

1

            Echogenicity: hypo- iso,or hyperechoic

2

            Calcifications: micro- or macrocalcifications

3

            Margins: well-defined/smooth or irregular

4

            Vascularity: high or low

5

            Shape: taller than wide

6

            Composition: Solid, Cystic, Mixed

 

 

Professional organizations such as the American Thyroid Association (51) and the American College of Radiology (50) developed ultrasound based risk stratification systems based on the above criteria and assigned a value or points to each of those when increasing the risk of malignancy, and assigning cutoff size when deciding FNA and to avoid unnecessary over diagnosis. There are several other Thyroid Imaging Reporting and Data Systems (TIRADS) and current efforts aim at developing an international TIRADS to unify lexicons and definitions and ultimately unify recommendations.

 

Neck ultrasound also plays a predominant role in the follow-up of thyroid nodules and thyroid cancer, including active surveillance of small papillary thyroid carcinomas.

 

An ever-increasing number of practicing endocrinologists are training to use US in routine practice. US is now used to supplement physical examination, when a thyroid mass is present, when a nodule needs careful measurement, or when an impalpable thyroid lesion is suspected. As a result of this widespread use, many small (<1.5 cm) thyroid incidentalomas are detected, creating what has been referred to as a “thyroid nodule epidemic” (3, 7, 53). Such a finding has been an unintended consequence of thyroid US use and has created a management dilemma for the clinicians.

 

The overall predictive value of US for malignancy based on a systematic review and meta-analysis of multiple studies is summarized in Table 6 (54). Although no single US feature is diagnostic for malignancy, the specificity is highest for taller-than-wide and microcalcifications, and lowest for echogenicity. The presence of at least 2 suspicious US criteria reliably identifies 85% to 93% of thyroid malignancies (3).

 

 

Table 6. Value of US Features Predicting Thyroid Malignancy*

US Feature

 

Sensitivity, %

 

Specificity, %

 

Microcalcifications

39.5

 

87.8

 

Hypoechogenicity

62.7

 

62.3

 

Irregular margins

50.5

 

83.1

 

Solid

72.7

 

53.2

 

Intranodular vascularity

45.9

 

78.0

 

Taller than wide

26.7

 

96.6

 

*Adapted and modified from Remonti et al. (54)

 

Sensitivity, positive predictive value, and negative predictive value increase significantly with US-FNA (3, 55-58). Nowadays, FNA should be exclusively performed with US-guidance. US-FNA permits precise needle placement in a nodule, thereby increasing both the rate of satisfactory aspirates and the diagnostic accuracy (3, 55-58).

 

FNA PITFALLS

 

The experience as well as the expertise of the cytopathologist is critical in avoiding pitfalls. Determining the adequacy of an aspirate, cellular atypia, performing and interpreting immunostains, and differentiation of lymphocytic thyroiditis from lymphoma are but a few of the challenges. Larger nodules are more likely to yield false‑negative results. To improve sampling, aspirates should be obtained from multiple sites of the nodule rather than repeatedly from a single spot. The absence of malignant cells in an otherwise acellular specimen does not exclude malignancy. It is good practice to biopsy all suspicious nodules in a multinodular gland (3, 4).

 

RE-BIOPSY

 

Opinions on indications for re-aspiration are divided, some favoring (11, 59), others not favoring (4, 60) routine re-biopsy. Lucas et al. (60) reported no advantage in routine re-biopsy, whereas Chehade et al. (59) found that repeated biopsy may decrease the rate of false-negative FNA from an average of 5.2% to less than 1.3%. Thyroid nodule guidelines from the American Association of Clinical Endocrinologists and the Associazione Medici Endocrinologi (4)did not suggest routine re-biopsy of FNA-benign nodules. The American Thyroid Association also recommends against routine re-biopsy (51). Repeat FNA for nodules that grow during serial sonographic follow-up can be done although nodule growth can be defined variably due to interobserver variation. Minimally significant change in nodule size of 50% increase in volume may warrant repeat FNA (61). Additional indications for re-biopsy are a non-diagnostic initial FNA, indeterminate cytology, and sonographically suspicious nodules with a benign cytology. In such instances, the current ATA guidelines recommend repeat biopsy within 1 year.

 

BETHESDA SYSTEM

 

The Bethesda System for reporting diagnostic criteria is shown in table 7 and the risk of malignancy for each diagnostic criteria in table 8.

 

 

Table 7. The Bethesda System for Reporting Thyroid Cytopathology: Recommended Diagnostic Categories

I.

Nondiagnostic or unsatisfactory

·       Cyst fluid only

·       Virtually acellular specimen

·       Other (obscuring blood, clotting artifact, etc.)

II.

Benign

·       Consistent with a benign follicular nodule (includes adenomatoid nodule, colloid nodule, etc.)

·       Consistent with lymphocytic thyroiditis (Hashimoto) in the proper clinical context

·       Consistent with granulomatous thyroiditis (subacute)

·       Other

III.

Atypia of undetermined significance or follicular lesion of undetermined significance

IV.

Follicular neoplasm or suspicious for a follicular neoplasm

·       Specify if Hürthle cell type (oncocytic)

V.

Suspicious for malignancy

·       Suspicious for papillary carcinoma

·       Suspicious for medullary carcinoma

·       Suspicious for metastatic carcinoma

·       Suspicious for lymphoma

·       Other

VI.

Malignant

·       Papillary thyroid carcinoma

·       Poorly differentiated carcinoma

·       Medullary thyroid carcinoma

·       Undifferentiated carcinoma (anaplastic)

·       Squamous cell carcinoma

·       Carcinoma with mixed features (specify)

·       Metastatic carcinoma

·       Non-Hodgkin lymphoma

·       Other

*Adapted with permission from Cibas and Ali (62).

 

 

Table 8. The Bethesda System for Reporting Thyroid Cytopathology: Risk of Malignancy and Recommended Clinical Management

Diagnostic Category

Risk of Malignancy (%)**

Usual Management

Nondiagnostic or unsatisfactory

5-10

Repeat FNA with ultrasound guidance

Benign            

0-3

Clinical follow-up

Atypia of undetermined significance or follicular lesion of undetermined significance                        

6-18

Repeat FNA, molecular testing, or lobectomy

Follicular neoplasm or suspicious for a follicular neoplasm                                         

10-40

Molecular testing, Lobectomy

Suspicious for malignancy

45-60

Near total thyroidectomy or lobectomy

Malignant       

94-96

Near total thyroidectomy or lobectomy

FNA-fine needle aspiration

*Adapted with permission from Cibas and Ali (62)

** When NIFTP is not considered cancer

 

 

THE UTILITY OF MOLECULAR TESTING IN THYROUD FNA/BIOPSY

 

Molecular testing is particularly useful in indeterminate cytology such as Bethesda class III or class IV that are expected to have a malignancy risk of 18 and 40% respectively (Table 8).

The use of molecular markers in indeterminate thyroid nodule cytology has lowered the need for diagnostic lobectomy in those cases. Repeat biopsy of Bethesda class III and IV can still be done to obtain more definitive diagnosis but the cytological assessment of a 2nd FNA only provide definitive diagnosis in about 40% (63).

 

Molecular testing of the thyroid FNA samples has significantly evolved since its introduction at the beginning of the 21st century. Multiple methods were developed for this approach and the tests are based on 3 main pathways: testing for somatic mutations, gene expression profiles, and microRNA (MiRNA) classifiers.

 

The most widely used molecular tests, ThyroSeq and Afirma assays have reasonable positive and negative predictive values suitable for using them when ruling-in and ruling-out thyroid cancer. Thyroseq V3 and Afirma GSC perform best to rule out malignancy (64)

 

Afirma molecular testing initially started as a microarray analysis of mRNA expression, currently a Genomic Sequencing Classifier (GSC) relying on RNA sequencing approach with a sensitivity of 96%, specificity of 68%, and positive predictive value of 47% (65).

 

The current version of ThyroSeq (version 3) is based on targeted next-generation sequencing analysis of 112 cancer related genes for point mutations, gene fusions, copy number alterations, and abnormal gene expression. This test has a reported sensitivity of 94% and a specificity of 82%, with a negative predictive value of 97%, and positive predictive value of 66% (66).

 

Other molecular tests may include a combination of more than one method such as those based on miRNA classification (ThyraMIR) and next-generation sequencing mutational analysis (ThyGeNEXT) which have excellent positive and negative predictive values (67).

 

When obtaining molecular testing, an additional dedicated needle pass can be useful. However, it is also possible to rinse the needles of all passes in the preservation fluid or cells from cytology slides. Furthermore, original cytology slides can be used in select cases such as in ThyGeNEXT/ThyraMIR (68).

 

THE USE OF US-GUIDED FNA WITH MINIMALLY INVASIVE THYROID PROCEDURES

 

Ultrasound-guided minimally invasive interventional techniques have been widely recognized and increasingly used over the last two decades. Those techniques include percutaneous ethanol injection (PEI) and thermal ablations such as laser and radiofrequency ablation. They can be used to treat both benign and malignant thyroid lesions. This includes the treatment of small papillary carcinomas, especially in patients who do not wish to pursue active surveillance, and in patients who are poor surgical candidates (69). Multiple international consensus statements were published to guide the use of these techniques with rapidly evolving indications (70-73).

 

Pre-procedural evaluation involves performing neck ultrasound, determining thyroid function status, and requiring two benign cytology for most solid nodules or one benign cytology for autonomously functioning thyroid nodules with low-risk thyroid ultrasound features (69).

 

PEI is used for predominantly cystic benign thyroid nodules. It was introduced in the early 1990s as successful treatment for benign thyroid nodules (74-76). With success rate defined as volume reduction of more than 50% with symptom control, PEI can reduce nodule volume from 50% to 98% (77, 78). Long-term outcomes of cystic nodule treated with PEI are excellent with minimum side effects and proven benefit. The procedure can be done in the outpatient setting under local anesthesia (69). Radiofrequency ablation and laser ablation can be also effectively used in the outpatient setting under local anesthesia for treating benign thyroid nodules, in particular autonomous nodules with a volume reduction range 50% to 85%, usually with better outcomes in smaller nodules (<10 mL) (69). Thermal ablation can also be performed in selective cases of thyroid malignancy such as low risk papillary thyroid microcarcinomas (79, 80). Complications include local pain, dysphonia, skin irritation, hematoma, and rarely nodule rupture (69).

 

 

REFERENCES

 

  1. Siegel RL, Miller KD, Jemal A 2020 Cancer statistics, 2020. CA Cancer J Clin 70:7-30.
  2. Gharib H 1994 Fine-needle aspiration biopsy of thyroid nodules: advantages, limitations, and effect. Mayo Clinic proceedings 69:44-49.
  3. Gharib H, Papini E 2007 Thyroid nodules: clinical importance, assessment, and treatment. Endocrinology and metabolism clinics of North America 36:707-735, vi.
  4. Gharib H, Papini E, Paschke R, Duick DS, Valcavi R, Hegedüs L, Vitti P 2010 American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and European Thyroid Association Medical guidelines for clinical practice for the diagnosis and management of thyroid nodules: executive summary of recommendations. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 16:468-475.
  5. Hegedüs L 2004 Clinical practice. The thyroid nodule. The New England journal of medicine 351:1764-1771.
  6. Mazzaferri EL 1993 Management of a solitary thyroid nodule. The New England journal of medicine 328:553-559.
  7. Tan GH, Gharib H 1997 Thyroid incidentalomas: management approaches to nonpalpable nodules discovered incidentally on thyroid imaging. Annals of internal medicine 126:226-231.
  8. Solomon BL, Wartofsky L, Burman KD 1996 Current trends in the management of well differentiated papillary thyroid carcinoma. The Journal of clinical endocrinology and metabolism 81:333-339.
  9. Martin HE, Ellis EB 1930 BIOPSY BY NEEDLE PUNCTURE AND ASPIRATION. Annals of surgery 92:169-181.
  10. Bäckdahl M, Wallin G, Löwhagen T, Auer G, Granberg PO 1987 Fine-needle biopsy cytology and DNA analysis. Their place in the evaluation and treatment of patients with thyroid neoplasms. The Surgical clinics of North America 67:197-211.
  11. Belfiore A, La Rosa GL 2001 Fine-needle aspiration biopsy of the thyroid. Endocrinology and metabolism clinics of North America 30:361-400.
  12. Atkinson BF 1993 Fine needle aspiration of the thyroid. Monographs in pathology:166-199.
  13. Goellner JR, Gharib H, Grant CS, Johnson DA 1987 Fine needle aspiration cytology of the thyroid, 1980 to 1986. Acta cytologica 31:587-590.
  14. Oertel YC 1996 Fine-needle aspiration and the diagnosis of thyroid cancer. Endocrinology and metabolism clinics of North America 25:69-91.
  15. Singer PA 1996 Evaluation and management of the solitary thyroid nodule. Otolaryngologic clinics of North America 29:577-591.
  16. Cao C, Jasim S, Cherian A, Nassar A, Chindris AM, Rivas AM, Bonnett S, Caserta M, Stan MN, Bernet VJ 2020 Patient Discomfort in Relation to Thyroid Nodule Fine-Needle Aspiration (FNA) Performed with or without Parenteral and/or Topical Anesthetic. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 26:1497-1504.
  17. Leboulleux S, Borget I, Labro S, Bidault S, Vielh P, Hartl D, Dauchy S, Chougnet CN, Girard E, Azoulay S, Mirghani H, Berdelou A, Lumbroso J, Deandreis D, Baudin E, Schlumberger M, Laurent S 2013 Frequency and intensity of pain related to thyroid nodule fine-needle aspiration cytology. Thyroid : official journal of the American Thyroid Association 23:1113-1118.
  18. Michael CW, Kameyama K, Kitagawa W, Azar N 2020 Rapid on-site evaluation (ROSE) for fine needle aspiration of thyroid: benefits, challenges and innovative solutions. Gland Surg 9:1708-1715.
  19. Hamburger JI 1994 Diagnosis of thyroid nodules by fine needle biopsy: use and abuse. The Journal of clinical endocrinology and metabolism 79:335-339.
  20. Lyle MA, Dean DS 2015 Ultrasound-guided fine-needle aspiration biopsy of thyroid nodules in patients taking novel oral anticoagulants. Thyroid : official journal of the American Thyroid Association 25:373-376.
  21. Khadra H, Kholmatov R, Monlezun D, Kandil E 2018 Do anticoagulation medications increase the risk of haematoma in ultrasound-guided fine needle aspiration of thyroid lesions? Cytopathology 29:565-568.
  22. Santos JE, Leiman G 1988 Nonaspiration fine needle cytology. Application of a new technique to nodular thyroid disease. Acta cytologica 32:353-356.
  23. Wang C, Vickery AL, Jr., Maloof F 1976 Needle biopsy of the thyroid. Surg Gynecol Obstet 143:365-368.
  24. Carpi A, Nicolini A, Sagripanti A, Righi C, Menchini Fabris F, Di Coscio G 2000 Large-needle aspiration biopsy for the preoperative selection of palpable thyroid nodules diagnosed by fine-needle aspiration as a microfollicular nodule or suspected cancer. American journal of clinical pathology 113:872-877.
  25. Hales MS, Hsu FS 1990 Needle tract implantation of papillary carcinoma of the thyroid following aspiration biopsy. Acta cytologica 34:801-804.
  26. Baloch ZW, LiVolsi VA 2004 Fine-needle aspiration of thyroid nodules: past, present, and future. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 10:234-241.
  27. Baloch ZW, Sack MJ, Yu GH, Livolsi VA, Gupta PK 1998 Fine-needle aspiration of thyroid: an institutional experience. Thyroid : official journal of the American Thyroid Association 8:565-569.
  28. Bisi H, de Camargo RY, Longatto Filho A 1992 Role of fine-needle aspiration cytology in the management of thyroid nodules: review of experience with 1,925 cases. Diagnostic cytopathology 8:504-510.
  29. Gharib H, Goellner JR, Johnson DA 1993 Fine-needle aspiration cytology of the thyroid. A 12-year experience with 11,000 biopsies. Clinics in laboratory medicine 13:699-709.
  30. Giuffrida D, Gharib H 1995 Controversies in the management of cold, hot, and occult thyroid nodules. The American journal of medicine 99:642-650.
  31. Reeve TS, Delbridge L, Sloan D, Crummer P 1986 The impact of fine-needle aspiration biopsy on surgery for single thyroid nodules. The Medical journal of Australia 145:308-311.
  32. Gharib H, Goellner JR 1993 Fine-needle aspiration biopsy of the thyroid: an appraisal. Annals of internal medicine 118:282-289.
  33. Caruso D, Mazzaferri EL 1991 Fine Needle Aspiration Biopsy in the Management of Thyroid Nodules. The Endocrinologist 1:194-202.
  34. Hall TL, Layfield LJ, Philippe A, Rosenthal DL 1989 Sources of diagnostic error in fine needle aspiration of the thyroid. Cancer 63:718-725.
  35. Gharib H, Goellner JR 1995 Fine-needle aspiration biopsy of thyroid nodules. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 1:410-417.
  36. Kini SR 1993 Needle aspiration biopsy of the thyroid: revisited. Diagnostic cytopathology 9:249-251.
  37. Tuttle RM, Lemar H, Burch HB 1998 Clinical features associated with an increased risk of thyroid malignancy in patients with follicular neoplasia by fine-needle aspiration. Thyroid : official journal of the American Thyroid Association 8:377-383.
  38. Schlinkert RT, van Heerden JA, Goellner JR, Gharib H, Smith SL, Rosales RF, Weaver AL 1997 Factors that predict malignant thyroid lesions when fine-needle aspiration is "suspicious for follicular neoplasm". Mayo Clinic proceedings 72:913-916.
  39. Baloch ZW, Fleisher S, LiVolsi VA, Gupta PK 2002 Diagnosis of "follicular neoplasm": a gray zone in thyroid fine-needle aspiration cytology. Diagnostic cytopathology 26:41-44.
  40. Gharib H 1997 Changing concepts in the diagnosis and management of thyroid nodules. Endocrinology and metabolism clinics of North America 26:777-800.
  41. MacDonald L, Yazdi HM 1996 Nondiagnostic fine needle aspiration biopsy of the thyroid gland: a diagnostic dilemma. Acta cytologica 40:423-428.
  42. McHenry CR, Walfish PG, Rosen IB 1993 Non-diagnostic fine needle aspiration biopsy: a dilemma in management of nodular thyroid disease. The American surgeon 59:415-419.
  43. Schmidt T, Riggs MW, Speights VO, Jr. 1997 Significance of nondiagnostic fine-needle aspiration of the thyroid. Southern medical journal 90:1183-1186.
  44. Chow LS, Gharib H, Goellner JR, van Heerden JA 2001 Nondiagnostic thyroid fine-needle aspiration cytology: management dilemmas. Thyroid : official journal of the American Thyroid Association 11:1147-1151.
  45. Alexander EK, Heering JP, Benson CB, Frates MC, Doubilet PM, Cibas ES, Marqusee E 2002 Assessment of nondiagnostic ultrasound-guided fine needle aspirations of thyroid nodules. The Journal of clinical endocrinology and metabolism 87:4924-4927.
  46. Samir AE, Vij A, Seale MK, Desai G, Halpern E, Faquin WC, Parangi S, Hahn PF, Daniels GH 2012 Ultrasound-guided percutaneous thyroid nodule core biopsy: clinical utility in patients with prior nondiagnostic fine-needle aspirate. Thyroid : official journal of the American Thyroid Association 22:461-467.
  47. Suh CH, Baek JH, Kim KW, Sung TY, Kim TY, Song DE, Choi YJ, Lee JH 2016 The Role of Core-Needle Biopsy for Thyroid Nodules with Initially Nondiagnostic Fine-Needle Aspiration Results: A Systematic Review and Meta-Analysis. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 22:679-688.
  48. Espinosa De Ycaza AE, Lowe KM, Dean DS, Castro MR, Fatourechi V, Ryder M, Morris JC, Stan MN 2016 Risk of Malignancy in Thyroid Nodules with Non-Diagnostic Fine-Needle Aspiration: A Retrospective Cohort Study. Thyroid : official journal of the American Thyroid Association 26:1598-1604.
  49. Gharib H, Papini E, Garber JR, Duick DS, Harrell RM, Hegedus L, Paschke R, Valcavi R, Vitti P, Nodules AAATFoT 2016 American Association of Clinical Endocrinologists, American College of Endocrinology, and Associazione Medici Endocrinologi Medical Guidelines for Clinical Practice for the Diagnosis and Management of Thyroid Nodules--2016 Update. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 22:622-639.
  50. Grant EG, Tessler FN, Hoang JK, Langer JE, Beland MD, Berland LL, Cronan JJ, Desser TS, Frates MC, Hamper UM, Middleton WD, Reading CC, Scoutt LM, Stavros AT, Teefey SA 2015 Thyroid Ultrasound Reporting Lexicon: White Paper of the ACR Thyroid Imaging, Reporting and Data System (TIRADS) Committee. J Am Coll Radiol 12:1272-1279.
  51. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L 2016 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid : official journal of the American Thyroid Association 26:1-133.
  52. Kobaly K, Kim CS, Mandel SJ 2022 Contemporary Management of Thyroid Nodules. Annu Rev Med 73:517-528.
  53. Ross DS 2002 Nonpalpable thyroid nodules--managing an epidemic. The Journal of clinical endocrinology and metabolism 87:1938-1940.
  54. Remonti LR, Kramer CK, Leitao CB, Pinto LC, Gross JL 2015 Thyroid ultrasound features and risk of carcinoma: a systematic review and meta-analysis of observational studies. Thyroid : official journal of the American Thyroid Association 25:538-550.
  55. Carmeci C, Jeffrey RB, McDougall IR, Nowels KW, Weigel RJ 1998 Ultrasound-guided fine-needle aspiration biopsy of thyroid masses. Thyroid : official journal of the American Thyroid Association 8:283-289.
  56. Cochand-Priollet B, Guillausseau PJ, Chagnon S, Hoang C, Guillausseau-Scholer C, Chanson P, Dahan H, Warnet A, Tran Ba Huy PT, Valleur P 1994 The diagnostic value of fine-needle aspiration biopsy under ultrasonography in nonfunctional thyroid nodules: a prospective study comparing cytologic and histologic findings. The American journal of medicine 97:152-157.
  57. Danese D, Sciacchitano S, Farsetti A, Andreoli M, Pontecorvi A 1998 Diagnostic accuracy of conventional versus sonography-guided fine-needle aspiration biopsy of thyroid nodules. Thyroid : official journal of the American Thyroid Association 8:15-21.
  58. Izquierdo R, Arekat MR, Knudson PE, Kartun KF, Khurana K, Kort K, Numann PJ 2006 Comparison of palpation-guided versus ultrasound-guided fine-needle aspiration biopsies of thyroid nodules in an outpatient endocrinology practice. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 12:609-614.
  59. Chehade JM, Silverberg AB, Kim J, Case C, Mooradian AD 2001 Role of repeated fine-needle aspiration of thyroid nodules with benign cytologic features. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 7:237-243.
  60. Lucas A, Llatjós M, Salinas I, Reverter J, Pizarro E, Sanmartí A 1995 Fine-needle aspiration cytology of benign nodular thyroid disease. Value of re-aspiration. European journal of endocrinology 132:677-680.
  61. Brauer VF, Eder P, Miehle K, Wiesner TD, Hasenclever H, Paschke R 2005 Interobserver variation for ultrasound determination of thyroid nodule volumes. Thyroid : official journal of the American Thyroid Association 15:1169-1175.
  62. Cibas ES, Ali SZ 2017 The 2017 Bethesda System for Reporting Thyroid Cytopathology. Thyroid : official journal of the American Thyroid Association 27:1341-1346.
  63. Allen L, Al Afif A, Rigby MH, Bullock MJ, Trites J, Taylor SM, Hart RD 2019 The role of repeat fine needle aspiration in managing indeterminate thyroid nodules. J Otolaryngol Head Neck Surg 48:16.
  64. Silaghi CA, Lozovanu V, Georgescu CE, Georgescu RD, Susman S, Nasui BA, Dobrean A, Silaghi H 2021 Thyroseq v3, Afirma GSC, and microRNA Panels Versus Previous Molecular Tests in the Preoperative Diagnosis of Indeterminate Thyroid Nodules: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 12:649522.
  65. Patel KN, Angell TE, Babiarz J, Barth NM, Blevins T, Duh QY, Ghossein RA, Harrell RM, Huang J, Kennedy GC, Kim SY, Kloos RT, LiVolsi VA, Randolph GW, Sadow PM, Shanik MH, Sosa JA, Traweek ST, Walsh PS, Whitney D, Yeh MW, Ladenson PW 2018 Performance of a Genomic Sequencing Classifier for the Preoperative Diagnosis of Cytologically Indeterminate Thyroid Nodules. JAMA Surg 153:817-824.
  66. Steward DL, Carty SE, Sippel RS, Yang SP, Sosa JA, Sipos JA, Figge JJ, Mandel S, Haugen BR, Burman KD, Baloch ZW, Lloyd RV, Seethala RR, Gooding WE, Chiosea SI, Gomes-Lima C, Ferris RL, Folek JM, Khawaja RA, Kundra P, Loh KS, Marshall CB, Mayson S, McCoy KL, Nga ME, Ngiam KY, Nikiforova MN, Poehls JL, Ringel MD, Yang H, Yip L, Nikiforov YE 2019 Performance of a Multigene Genomic Classifier in Thyroid Nodules With Indeterminate Cytology: A Prospective Blinded Multicenter Study. JAMA Oncol 5:204-212.
  67. Labourier E, Shifrin A, Busseniers AE, Lupo MA, Manganelli ML, Andruss B, Wylie D, Beaudenon-Huibregtse S 2015 Molecular Testing for miRNA, mRNA, and DNA on Fine-Needle Aspiration Improves the Preoperative Diagnosis of Thyroid Nodules With Indeterminate Cytology. The Journal of clinical endocrinology and metabolism 100:2743-2750.
  68. Grani G, Sponziello M, Pecce V, Ramundo V, Durante C 2020 Contemporary Thyroid Nodule Evaluation and Management. The Journal of clinical endocrinology and metabolism 105:2869-2883.
  69. Jasim S, Patel KN, Randolph G, Adams S, Cesareo R, Condon E, Henrichsen T, Itani M, Papaleontiou M, Rangel L, Schmitz J, Stan MN 2022 American Association of Clinical Endocrinology Disease State Clinical Review: The Clinical Utility of Minimally Invasive Interventional Procedures in the Management of Benign and Malignant Thyroid Lesions. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 28:433-448.
  70. Garberoglio R, Aliberti C, Appetecchia M, Attard M, Boccuzzi G, Boraso F, Borretta G, Caruso G, Deandrea M, Freddi M, Gallone G, Gandini G, Gasparri G, Gazzera C, Ghigo E, Grosso M, Limone P, Maccario M, Mansi L, Mormile A, Nasi PG, Orlandi F, Pacchioni D, Pacella CM, Palestini N, Papini E, Pelizzo MR, Piotto A, Rago T, Riganti F, Rosato L, Rossetto R, Scarmozzino A, Spiezia S, Testori O, Valcavi R, Veltri A, Vitti P, Zingrillo M 2015 Radiofrequency ablation for thyroid nodules: which indications? The first Italian opinion statement. J Ultrasound 18:423-430.
  71. Hahn SY, Shin JH, Na DG, Ha EJ, Ahn HS, Lim HK, Lee JH, Park JS, Kim JH, Sung JY, Lee JH, Baek JH, Yoon JH, Sim JS, Lee KH, Baek SM, Jung SL, Kim YK, Choi YJ, Korean Society of Thyroid R, Korean Society of R 2019 Ethanol Ablation of the Thyroid Nodules: 2018 Consensus Statement by the Korean Society of Thyroid Radiology. Korean J Radiol 20:609-620.
  72. Kim JH, Baek JH, Lim HK, Ahn HS, Baek SM, Choi YJ, Choi YJ, Chung SR, Ha EJ, Hahn SY, Jung SL, Kim DS, Kim SJ, Kim YK, Lee CY, Lee JH, Lee KH, Lee YH, Park JS, Park H, Shin JH, Suh CH, Sung JY, Sim JS, Youn I, Choi M, Na DG, Guideline Committee for the Korean Society of Thyroid R, Korean Society of R 2018 2017 Thyroid Radiofrequency Ablation Guideline: Korean Society of Thyroid Radiology. Korean J Radiol 19:632-655.
  73. Orloff LA, Noel JE, Stack BC, Jr., Russell MD, Angelos P, Baek JH, Brumund KT, Chiang FY, Cunnane MB, Davies L, Frasoldati A, Feng AY, Hegedus L, Iwata AJ, Kandil E, Kuo J, Lombardi C, Lupo M, Maia AL, McIver B, Na DG, Novizio R, Papini E, Patel KN, Rangel L, Russell JO, Shin J, Shindo M, Shonka DC, Jr., Karcioglu AS, Sinclair C, Singer M, Spiezia S, Steck JH, Steward D, Tae K, Tolley N, Valcavi R, Tufano RP, Tuttle RM, Volpi E, Wu CW, Abdelhamid Ahmed AH, Randolph GW 2022 Radiofrequency ablation and related ultrasound-guided ablation technologies for treatment of benign and malignant thyroid disease: An international multidisciplinary consensus statement of the American Head and Neck Society Endocrine Surgery Section with the Asia Pacific Society of Thyroid Surgery, Associazione Medici Endocrinologi, British Association of Endocrine and Thyroid Surgeons, European Thyroid Association, Italian Society of Endocrine Surgery Units, Korean Society of Thyroid Radiology, Latin American Thyroid Society, and Thyroid Nodules Therapies Association. Head Neck 44:633-660.
  74. Lippi F, Ferrari C, Manetti L, Rago T, Santini F, Monzani F, Bellitti P, Papini E, Busnardo B, Angelini F, Pinchera A 1996 Treatment of solitary autonomous thyroid nodules by percutaneous ethanol injection: results of an Italian multicenter study. The Multicenter Study Group. The Journal of clinical endocrinology and metabolism 81:3261-3264.
  75. Monzani F, Caraccio N, Goletti O, Casolaro A, Lippolis PV, Cavina E, Miccoli P 1998 Treatment of hyperfunctioning thyroid nodules with percutaneous ethanol injection: Eight years' experience. Exp Clin Endocrinol Diabetes 106 Suppl 4:S54-58.
  76. Tarantino L, Francica G, Sordelli I, Sperlongano P, Parmeggiani D, Ripa C, Parmeggiani U 2008 Percutaneous ethanol injection of hyperfunctioning thyroid nodules: long-term follow-up in 125 patients. AJR Am J Roentgenol 190:800-808.
  77. Bennedbaek FN, Nielsen LK, Hegedüs L 1998 Effect of percutaneous ethanol injection therapy versus suppressive doses of L-thyroxine on benign solitary solid cold thyroid nodules: a randomized trial. The Journal of clinical endocrinology and metabolism 83:830-835.
  78. Guglielmi R, Pacella CM, Bianchini A, Bizzarri G, Rinaldi R, Graziano FM, Petrucci L, Toscano V, Palma E, Poggi M, Papini E 2004 Percutaneous ethanol injection treatment in benign thyroid lesions: role and efficacy. Thyroid : official journal of the American Thyroid Association 14:125-131.
  79. Choi Y, Jung SL 2020 Efficacy and Safety of Thermal Ablation Techniques for the Treatment of Primary Papillary Thyroid Microcarcinoma: A Systematic Review and Meta-Analysis. Thyroid : official journal of the American Thyroid Association 30:720-731.
  80. Yan L, Zhang M, Song Q, Luo Y 2021 Ultrasound-Guided Radiofrequency Ablation Versus Thyroid Lobectomy for Low-Risk Papillary Thyroid Microcarcinoma: A Propensity-Matched Cohort Study of 884 Patients. Thyroid : official journal of the American Thyroid Association 31:1662-1672.

 

 

Testicular Cancer: Pathogenesis, Diagnosis and Management with Focus on Endocrine Aspects

ABSTRACT

 

Testicular cancer comprises different neoplasms, depending on the cell of origin and the typical age at presentation, but germ cell-derived tumors constitute the vast majority of cases. Testicular germ cell tumors (TGCT) can be diagnosed in every age group, but more than 90% of cases occur in young men. These tumors, comprising seminoma and nonseminoma, are derived from germ cell neoplasia in situ (GCNIS). Pathogenesis of TGCT associated with GCNIS partly overlaps with that of other developmental disorders of the male reproductive system within the testicular dysgenesis syndrome (TDS). Testicular somatic cell neoplasms, known as sex cord-stromal tumors, are relatively rare, but can have endocrine manifestations, such as precocious puberty or gynecomastia. In addition to its malignant features, cancer of the testis represents a developmental, endocrine, and reproductive problem. These issues are the focus of this chapter, and emphasis is given to aspects that are of interest to endocrinologists, including pediatric endocrinologists and andrologists. Management of invasive testicular tumors is largely handled by urologists and oncologists, thus only general information on surgical treatment, radiotherapy, and chemotherapy is presented. Impact of cancer treatment on the endocrine system, co-morbidities, fertility issues, and quality of life issues are also briefly reviewed.

 

INTRODUCTION

                                                           

Testicular cancer comprises a number of different neoplasms, depending on the cell of origin and the typical age at presentation (1, 2). Although several cell types in the testis can undergo neoplastic transformation, germ cell-derived tumors constitute the vast majority of cases of testicular neoplasms. The relative distribution depends on age: in young adult men nearly all tumors are germ cell tumors, whereas in patients aged 70 years or older, a large proportion of lymphomas and secondary carcinomas can be expected (2).  In other words, as explained in detail further in the text, germ cell cancer can be diagnosed in every age group, but more than 90% of cases occur in young men, and this subgroup is the main focus of this chapter. Pathogenesis of testicular germ cell tumors (TGCT) of young adults partly overlaps with that of other developmental disorders of the male reproductive system, within the testicular dysgenesis syndrome (TDS) (3, 4, 5). Somatic cell tumors in the testis, known as sex cord-stromal neoplasms are relatively rare, but are also discussed in this chapter. These neoplasms can have endocrine manifestations due to their origin from endocrine cells.

 

In addition to its malignant features, cancer of the testis represents a developmental, endocrine, and reproductive problem. These issues are the focus of this chapter, and emphasis is given to aspects that are of interest to endocrinologists, including pediatric endocrinologists and andrologists. The contribution of andrologists and endocrinologists to the overall management of patients with testicular cancer is very important, especially concerning early diagnosis, fertility issues, testosterone deficiency, and the impact of treatment on the quality of life of the patients, the majority of whom are young adults. We have to emphasize that only very general information on surgical and oncological treatment options (e.g., chemotherapy) is presented here. However, urologists and oncologists, who are responsible for the clinical management of testicular tumors, would also benefit from learning about the pathogenesis and andrological aspects of testicular cancer summarized in this chapter.

 

GERM CELL TUMORS (GCT)

 

Germ cell tumors (GCT) are characterized by extreme phenotypic heterogeneity (2). They display features of pluripotency and about half of them (nonseminomas) can differentiate into virtually any somatic tissue type within the so-called teratomas that recapitulate early embryonic differentiation (6, 7, 8). Although in comparison to other solid tissue cancers, GCTs are relatively rare, they constitute the most common form of solid tissue cancer of young age. The typical localization of GCT is the testis in males and ovary in females, but GCTs may also be found outside the gonads, thus these tumors are named extragonadal GCTs (8, 9).  Extragonadal GCTs occur most often in children of both genders, preferentially along the body midline (intracranial, pineal, mediastinal) but can occur also in adults (8, 9). An increased frequency of extragonadal GCTs, especially in mediastinum, has been associated with Klinefelter syndrome (10). It is important to remember that a large proportion of cases of extragonadal GCT in retroperitoneal locations are associated with pre-malignant changes in testicles, and can be assumed early metastases of testicular neoplasms, although a multi-site development of GCT cannot be completely excluded (8, 9, 11, 12). This chapter focuses on the testicular GCT. For the extragonadal GCT in children and adults, the reader should consult specialized literature.

 

Classification and Histopathology of Testicular Germ Cell Tumors (TGCT)

 

Testicular germ cell tumors (TGCT) are by far the most frequent neoplasms of the testis and comprise approximately 90-95% of cases. They may affect infants (rarely), young men (commonly) and elderly men (rarely). The TGCTs of childhood are mentioned only briefly at the end of this section. The diagnosis and management aspects of TGCT in adult men is the focus of this chapter.

 

The most commonly accepted and currently used classification is the WHO classification of testicular tumors. The main changes occurred in 2016 when the WHO consensus panel proposed a thorough revision of the classification, which for the first time was based on biological evidence (2). The most radical change was the new division of TGCT into two major groups according to the origin from germ cell neoplasia in situ (GCNIS), and coining the term GCNIS, replacing several previously used and confusing names (2, 13). The latest 2022 WHO classification update proposed only minor adjustments; the most important is recognition of gonadoblastoma as a precursor similar to GCNIS and proposing placing seminoma under the major category of germinomas (14).

 

For use by non-pathologists, a simplified division of TGCT shown in Table 1.

 

Table 1. Simplified Classification of Testicular Germ Cell Tumors (TGCT), Based on the WHO Classification.

·                GCT derived from germ cell neoplasia in situ (GCNIS)

o                 Non-invasive lesions: GCNIS (9064/2)* and gonadoblastoma (9073/1)

o                 Germinomas:

-                                                    Seminoma, pure (9061/3)

-                                                    Seminoma with syncythiotrophoblastic cells

o                 Nonseminomatous (non-germinomatous) GCT, pure

-                                                    Embryonal carcinoma (9070/3)

-                                                    Yolk sac tumor, postpubertal type (9071/3)

-                                                    Trophoblastic tumors, incl. choriocarcinoma (9100/3)

-                                                    Teratoma, postpubertal type (9080/3), incl. teratoma with somatic-type transformation (9084/3)

o                 Nonseminomatous (non-germinomatous) mixed germ cell tumors (9085/3)

o                 Regressed GCTs (9080/1)

·                GCT unrelated to GCNIS

o                 Spermatocytic tumor (9063/3)

o                 Prepubertal (pediatric) tumors

-                                                    Teratoma, prepubertal type (9084/0)

§                    Dermoid cyst

§                    Epidermoid cyst

-                                                    Yolk sac tumor, prepubertal type (9071/3)

-                                                    Prepubertal type testicular neuroendocrine tumor (8240/3)

-                                                    Mixed prepubertal type tumors (9085/3)

*Footnote: The codes in parentheses are from the International Classification of Diseases for Oncology (ICD-O-3).

 

Precursor Lesions

 

The TGCT of young adults originate from a common precursor, germ cell neoplasia in situ (GCNIS), initially termed carcinoma in situ (CIS) testis (13, 15). GCNIS is considered to originate from developmentally arrested immature germ cells (gonocytes) that fail to differentiate to spermatogonia (4, 16, 17). Accordingly, morphology of GCNIS cells resembles closely that of fetal gonocytes, but with more irregular chromatin in the nuclei. GCNIS cells are located inside seminiferous tubules, most frequently in a single row along the basement membrane (Figure 1).

 

Gonadoblastoma is a preinvasive lesion which occurs almost exclusively in individuals with disorders of sexual development (DSD). This lesion is most often found in female patients with mixed gonadal dysgenesis (45,X/46,XY) or Turner Syndrome but can also occur in males (2, 18, 19). Gonadoblastoma cells and GCNIS cells have a very similar gonocyte/oogonium-like phenotype, but the surrounding somatic cells are different; GCNIS cells are present inside seminiferous tubules, which are usually well developed but may be hypoplastic and contain immature Sertoli cells, while gonadoblastoma consists of groups of germ cells, which are nested in small stromal cells similar to granulosa cells (19, 20, 21, 22). GCNIS and gonadoblastoma can be present in the same gonad and there are also lesions with morphology in between the two entities (18, 19). The clinical course of pure gonadoblastoma may be benign, but it has a potential to transform into a malignant germ cell tumor, especially if accompanied by GCNIS and greater virilization of the patient (19, 22, 23, 24). 

Figure 1. Histology of germ cell neoplasia in situ (GCNIS). The upper panel (hematoxyllin-eosin, HE staining shows a low magnification view of GCNIS in a typical pattern with only GCNIS cells and Sertoli cells present inside tubules. The tubules with neoplasia have a smaller diameter than normal seminiferous tubules. On the right side of this image a few tubules with decreased spermatogenesis are visible. The lower left image shows a fragment of a tubule with GCNIS side-by-side with a tubule with preserved spermatogenesis; note the large GCNIS nuclei. The lower right image displays GCNIS cells visualized by immunohistochemical staining for placental-like alkaline phosphatase (PLAP).

 

The immunohistochemical profiles of GCNIS and gonadoblastoma cells are virtually identical and resemble very closely those of primordial germ cells and fetal gonocytes (21, 25, 26).  This was subsequently confirmed by comparative studies at the transcriptional level using microarrays (17, 27, 28).  Among many genes highly expressed in GCNIS cells (as well as gonadoblastoma, normal fetal germ cells and several TGCTs) the following should be mentioned because of their interesting biological function (germ cell survival or maintenance of embryonic stem cell pluripotency) and usefulness in histopathological diagnosis (24): OCT4 (29), AP2-gamma/TFAP2C (27, 30), NANOG (27, 31, 32), KIT (25, 33), TP53 (34), and LIN28 (35, 36).

 

In addition to protein-coding genes, GCNIS cells display a specific profile of embryonic-type micro-RNAs (miRNA); miR-371-3 cluster, miR-302 and miR-367 (37, 38). This miRNA profile is also detectable in overt TGCT, except teratoma (38), see the description in the section on serum markers. 

 

Testicular Germ Cell Tumors (TGCT) Derived From GCNIS

As mentioned in the introduction, TGCT of young adult men display very variable histology (some examples are shown in Figure 2) and are divided into seminomas (under the main GCT category of germinomas which occur in men and women) and nonseminomas (non-germinomas) (2, 14, 39).

Figure 2. Histology of main types of testicular germ cell tumors. The large images show a general histology pattern of a seminoma (upper panel) and two most often seen types of nonseminoma: undifferentiated embryonal carcinoma (middle panel) and teratoma, a tumor displaying differentiation into various somatic tissues (bottom panel). Small square pictures on the right show cellular characteristics in a greater magnification.  All sections are stained with hematoxylin-eosin (HE).

 

Seminomas are most often diagnosed in the 25- to 40-year-old age group, whereas nonseminomatous tumors occur in even younger men (adolescence to 30 years). Both types originate from GCNIS (2, 16). Seminoma resembles a mass of immature germ cells; the tumor cells are morphologically very close to GCNIS cells and proliferate as a homogeneous tumor, which retains features of germinal lineage. The gene expression profile of seminoma is similar to that of GCNIS and fetal gonocytes, and virtually identical to the female equivalent of germinoma, called dysgerminoma (26, 39).

 

Nonseminomatous tumors display a variety of histological forms and contain undifferentiated embryonal carcinoma and somatic components partly differentiated along embryonic lineage of any tissue type (1, 2, 8). Nonseminomas also contain extra-embryonic tissue components (yolk sac tumor and choriocarcinoma). The combined or mixed tumors contain elements of seminoma and nonseminoma but are classified and clinically treated as nonseminoma, which usually has more severe clinical course than seminoma (1, 2).

 

Testicular Germ Cell Tumors Not Associated with GCNIS

 

PREPUBERTAL (PEDIATRIC) TGCTs. 

 

These tumors occur in early childhood (between birth and approximately 5 years of age) and comprise two main histological types: yolk sac tumor of the prepubertal type and mature teratoma (including dermoid cyst) (Table 1). Prepubertal teratomas can contain secondary neuroendocrine elements, which can overgrow the tumor, hence, the latest WHO classification added this entity as the third subtype (14). The histology of the prepubertal tumors does not differ from the adult equivalents, which are components of nonseminoma (2).  Likewise, these tumors display similar characteristic transcriptome and micro-RNA profiles (40, 41). In contrast to the TGCT derived from GCNIS, the tumor genome does not display isochromosome 12p. The etiology and pathogenesis of infantile TGCT remain unknown. These tumors are assumed to originate from primordial germ cells (PGC) but there is no known precursor lesion of GCNIS/gonadoblastoma type, neither are there signs of testicular dysgenesis (2, 42, 43).

 

SPERMATOCYTIC TUMOR  

 

This rare tumor occurs primarily in older men (median age at diagnosis around 50-55 years, range 25-94) but occasionally can be diagnosed in men in in the third decade of life (44). Spermatocytic tumor has no extragonadal or ovarian counterpart, and occurs exclusively in post-pubertal testis (1, 2, 8). This tumor is not derived from GCNIS (45) and has the expression profile similar to spermatogonia B or early primary spermatocytes (26, 46, 47, 48). Spermatocytic tumors appear to grow from clonally expanding germ cells, which are committed to but have not yet entered meiosis. The following de novo genetic aberrations causing increased spermatogonial proliferation have been identified; amplifications in chromosome 9p encompassing DMRT1 locus (46), rare gain-of-function mutations in genes encoding FGFR2, FGFR3, HRAS, NRAS, PTPN11, and simultaneous whole chromosome gains of chromosome 9 and chromosome 20 combined with a loss of chromosome 7 (49, 50). Some of these mutations that occur spontaneously in germ cells of aging men have been depicted as “selfish mutations” because - if transmitted to next generation - can cause severe inborn skeletal abnormalities in the offspring, such as achondroplasia, hypochondroplasia, thanatophoric dysplasia or Costello syndrome (51).

 

Etiology and Pathogenesis of Testicular Germ Cell Cancer of Young Adults (Derived From GCNIS)

 

 

Epidemiology of testicular germ cell tumors (TGCT) has attracted the growing attention of researchers, because of the steadily rising incidence (51). The incidence is remarkably variable geographically and dependent on the ethnic background (Figure 3) (52). TGCT is currently the most common malignancy of young men of white European ancestry, while it is relatively rare among Asians and Africans (52, 53, 54). Interestingly, indigenous Maoris of New Zealand have a higher incidence of testicular cancer than the white population (55).

Figure 3. Global incidence rates of testicular cancer.  Age-standardized incidence rates for testicular cancer for men of all ages in selected countries of all regions of the world, extracted from national registries. National reporting systems varied by country, and data quality may have fluctuated between regions. Reprinted from Znaor et al., Eur Urol 2014 (52).

 

However, the incidence rates are not stable and have been changing over time. Within populations rapid changes have been observed in recent decades. For example, the highest rates of testicular cancer have historically been observed in Norway and Denmark but much lower in the Eastern and Southern Europe. While the incidence in Denmark has been attenuating since the 1970s, an alarming rise has been noted in Finland and in Slavic Balkan countries, especially Slovenia and Croatia (52, 53, 54, 56). Similarly, a rapid increase has been noted among the Hispanic/Latin population in the United States (57). Another interesting feature of epidemiology of testicular cancer is the so-called birth cohort effect; the rise in the incidence correlated with the calendar year of birth rather than with the age at diagnosis (58). For unexplained reasons, cohorts born at wartime had lower incidence than men born just before or after the war (58).

As mentioned before, testicular cancer has an unusual age-specific incidence rate with a small peak in the postnatal period and a major peak in young adult age, starting at puberty. These periods coincide with an activation of gonadal hormones, indicating there may be a possible connection between the hormonal factors and invasive transformation of germ cells. In certain risk groups, the incidence of testicular cancer is much increased. Individuals with developmental abnormalities of the gonads and sex differentiation (DSD) are at high risk of developing germ cell neoplasia. Among these, individuals with the so-called mixed gonadal dysgenesis and the 45,X/46,XY karyotype are at particular risk of harboring GCNIS or gonadoblastoma and developing TGCT in adolescence (18, 19, 20, 23, 59, 60). An association with testicular cancer has been noted in a number of developmental abnormalities, such as cryptorchidism (61, 62), Down syndrome (63) but also low birth weight and unspecific perinatal factors, e.g., premature birth, birth order, high levels of maternal estrogens or bleeding during pregnancy, high maternal age and neonatal jaundice (62, 64). A late age at puberty and tall stature were also associated with lower and higher risk of testicular cancer, respectively (65, 66).

 

For endocrinologists and andrologists it is essential to know that male infertility (the type linked to TDS) is one of the commonest risk factors for testicular cancer (67). Men with TGCT have poorer semen quality and significantly fewer children than controls prior to development of their tumor (68, 69), see also the section on TDS below.

 

Patients with a unilateral TGCT are also at an increased risk to develop a new primary testicular tumor of the contralateral testis. Depending on the studied population and the interval from the primary diagnosis, approximately 2 - 5% patients will be diagnosed with a second TGCT during their lifetime (70, 71, 72, 73). The majority of bilateral TGCT occur metachronously (> 6 months after the primary TGCT), some as late as 20-40 years after the primary TGCT (74). The histology of the primary tumor influences the risk only marginally, with the predominance of seminomas among the synchronous TGCTs but primary nonseminomas observed most often in younger patients with metachronous tumors developing after shorter intervals (70, 72). The presence of testicular atrophy increases the risk of bilateral neoplasia considerably (75, 76).

 

Testicular Dysgenesis Syndrome (TDS)

 

The association of testicular cancer with poor testicular function, cryptorchidism, hypospadias, and abnormal testicular development led to a hypothesis that poor gonadal development and testicular neoplasia are etiologically linked. A concept of TDS was proposed, in which testicular cancer is one of the symptoms, in addition to other phenotypes, including cryptorchidism, hypospadias, shortened anogenital distance (AGD), reduced Leydig cell function, and decreased spermatogenesis (3, 5). This hypothesis is supported by histological studies showing that dysgenetic features, such as undifferentiated tubules with visibly immature Sertoli cells, clusters of poorly differentiated tubules, or hyaline bodies, often seen in association with testicular cancer, are not uncommon among men referred to andrology clinics because of fertility problems (3, 5, 18, 71). It is important to underline that not all cases of genital malformations and infertility are a part of TDS. Milder phenotypes linked to impaired development of the testis in fetal life due to diverse environmental or lifestyle-related factors can be considered part of TDS (5, 77). Severe genital malformations caused by genetic disorders are clearly a part of DSD but there is a partial overlap between the two syndromes (18). However, TDS severity and prevalence are undoubtedly modulated by the genetic variability (polymorphisms) and epigenetics (5). A schematic representation of the pathogenesis and manifestations of TDS is depicted in Figure 4.

Figure 4. Schematic illustration of aetiology and pathogenesis of disorders grouped within Testicular Dysgenesis Syndrome (TDS). The TDS concept implicates disturbed function of testicular somatic cells (Leydig- and Sertoli cells) caused by inherited genetic variation (gene polymorphisms) in combination with environmental /lifestyle factors acting during early development. Dysfunction of the somatic cells results in disturbed hormonal homeostasis and causes impaired germ cell differentiation. Depending on the severity of the impairment, multiple outcomes or phenotypes may occur, ranging from reduced anogenital distance (AGD), genital malformations to testicular cancer. Note that the most severe forms of TDS (disorders of sex development with gonadoblastoma (GDB) or GCNIS are the least frequently seen, whereas the mildest forms, such as impaired spermatogenesis are quite common. Modified from Skakkebæk et al., Hum Reprod 2001 (3) and Physiol Rev 2016 (5).

 

Genomics of TGCT and Predisposing Polymorphisms  

 

Tumors derived from germ cells via GCNIS stage are characterized by the presence of a nearly universal aneuploidy (polyploidization) and amplification of chromosome 12p, often in the form of an isochromosome (8, 78). Additional secondary genomic aberrations in invasive TGCT include rare recurring chromosomal gains or losses (in particular loss of chromosome Y), and fusion transcripts (79, 80). Strikingly few oncogenic mutations have been reported in TGCTs, and the short list of affected genes includes only KIT (predominantly in seminomas) and KRAS (mainly in nonseminomas) (79, 81, 83).

 

Although testicular cancer in most cases is a sporadic disease, the familial risk of testicular GCT is among the highest when compared to other cancers; brothers and sons of TGCT cases have an 8-10-fold and 4-6-fold increased risk, respectively (84). A twin study from Nordic countries estimated the heritability of testicular cancer as 37%, with 24% attributed to shared environment (85). Recent studies using next generation sequencing technology confirmed the absence of specific oncogenic driver mutations that would explain a high heritable risk of testicular cancer (86). The bulk of inherited risk is instead caused by a constellation of unfavorable gene variants, which jointly account for estimated 44% of testicular cancer heritability (87). Men with a polygenic risk score in the 95th percentile have a 6.8-fold increased risk of TGCT compared to men with median scores (87). This estimation is based on numerous genome-wide association studies (GWAS) performed in recent decades, which revealed a large number of significant gene variants (markers) associated with TGCT risk (87, 88, 89, 90, 91, 92, 93). The strongest association, which was identified in all GWAS, and most interesting from the biological point of view, is with a cluster of single nucleotide polymorphisms (SNPs) within or near KIT ligand gene (KITLG) and in genes downstream of the KIT/KITLG/MAPK signaling pathway, e.g., SPRY4. This pathway is essential for germ cell migration and survival, is highly active in GCNIS, with the frequent secondary gain-of-function mutations identified in seminomas (33, 81, 82, 83, 89, 94, 95). Among other significant SNPs associated with TGCT risk, the most interesting are DMRT1, a transcription factor involved in sex differentiation and regulation of meiosis (96, 97), PRDM14 and DAZL, factors involved in primordial germ cell specification and differentiation, AMHR2, the receptor for anti-Mullerian hormone (AMH), AR, the androgen receptor, and several genes in the telomerase and DNA repair pathway (87, 90, 91, 92, 93). It is biologically important that several predisposing variants are potentially associated with germ cell differentiation and testis development, including hormone regulation, and there is an overlap with some pathways involved in TDS. 

Current Views on the Environmental Etiology of TGCT

In spite of significant recent inroads into the understanding of the pathogenesis of TGCT, etiology of these tumors, and especially the reason for the rise in incidence, remains obscure. The high incidence of testicular cancer in subjects with congenital errors of gonadal development strongly implicates the involvement of intrauterine factors and perinatal factors. We believe that the neoplastic transformation of male germ cells occurs during their pre-meiotic development, and this happens preferentially in individuals with genetic susceptibility. GCNIS cells and primordial germ cells share some distinct features, such as expression of embryonic pluripotency factors, low DNA methylation and constellation of miRNAs and histone modifications (4, 5, 17, 27, 28, 37, 39, 99, 100).

 

The mechanisms of neoplastic transformation of early germ cell are not known. There is a growing consensus that there may be multiple mechanisms and testicular cancer is a multifactorial and polygenic disorder. A disturbance in the fetal programming of gonadal development may be a result of an intrauterine hormonal imbalance, which in turn may be caused by a genetic disorder or by an impact of an exogenous factor targeting a key pathway, e.g., androgen signaling, KIT-KITLG signaling, DMRT1 signaling and regulation of meiosis, the TGF-beta superfamily regulation (including Nodal pathway) or the WNT pathway, leading to a delay in the testis development and maturation of fetal gonocytes (3, 4, 91, 98, 94, 101, 102, 103).

 

As mentioned above, the rising and quickly changing incidence of testicular cancer in well-developed countries suggests a possible adverse influence of environmental or lifestyle-related causative factors. In recent decades a great number of potent natural and synthetic hormones and hormone antagonists have been identified in environment. Observations in wildlife and experiments in laboratory animals exposed to synthetic hormones and a broad range of endocrine disrupters suggest that these substances can cause a disturbance of hormonal milieu of the developing gonad and disturb differentiation of early germ cells (3, 4, 5, 104, 105, 106).  Whether or not the endocrine disrupters have contributed to the rise in testicular cancer remains to be demonstrated. Human exposure studies are difficult, mainly because of the long interval between the vulnerable period of early development and manifestation of cancer development in adulthood. There is also the problem of the plethora of confounding factors, including mixtures of possible contaminating factors present in food, packaging, cosmetics, and other products of daily life. Therefore, the evidence for the role of endocrine disrupters in the etiology of TGCT remains scarce. Among few existing data one can mention a higher serum level of some persistent organic pollutants found in mothers of men with testicular cancer (107), and a greater burden of p,p′-DDE (a DDT metabolite) in serum of testicular cancer patients (108). For more information on the existing evidence and possible mechanisms the reader is referred to recent review articles on the subject (109, 110). It is clear that further studies in large cohorts of patients and controls are needed to identify the causative factors behind the observed testicular cancer epidemics. Few data exist concerning postnatal exposures and risk of TGCT. A heavy use of marijuana (cannabis) has been consistently reported as a risk factor for nonseminoma but the mechanism remains to be elucidated (111, 112).

 

Diagnosis and Staging of Testicular Germ Cell Neoplasms

PREINVASIVE LESIONS AND TESTICULAR BIOPSY


All attending physicians should consider a possibility of incipient germ cell neoplasia in men with infertility, especially those with small testes, a history of cryptorchidism, or previous testicular tumors, or in individuals from the high-risk group of DSD (5, 68, 69). The preinvasive stage of GCNIS is asymptomatic, so diagnosis at this early stage is sporadic. Ultrasonographic examination can be helpful in some cases. GCNIS or incipient TGCT are frequently associated with an irregular echo pattern or microcalcifications /microlithiasis (113, 114, 115, 116). However, ultrasonic microlithiasis is very common and can be present in normal men or patients with mild disorders, so it alone is not an indication for a testis biopsy. A biopsy is indicated if microlithiasis is accompanied by additional risk factors, e.g., history of cryptorchidism, a family history of testicular cancer, atrophic testis (<12 mL), or poor semen quality, especially azoospermia (117, 118). Surgical testicular biopsy is currently the only sure diagnostic procedure for GCNIS diagnosis (119). Experience in the use of needle biopsies for GCNIS diagnosis is limited and this method is not commonly used in the clinic. Regardless of the method of obtaining the testis tissue, examination for GCNIS by immunohistochemistry in a diagnostic biopsy or any leftover material after testicular sperm extraction in fertility clinics is now considered mandatory (119, 120, 121).

 

GCNIS is present in the testis contralateral to a testicular tumor in approximately 5% of cases, with a range of 4% to 8%, depending on the studied cohort (71, 75, 121, 122). Screening for the contralateral GCNIS to prevent the appearance of metachronous bilateral TGCT has been practiced in several countries, including nationwide in Denmark and in some centers in other European countries (Germany, Austria, the Netherlands). Most of other centers adopted a recommendation of the European Association of Urology (123) to take contralateral biopsies only in patients at risk for GCNIS presence, such as by a history of cryptorchidism, a small testis volume (<12 ml), or testicular microlithiasis (75, 76). In the USA and Canada, the contralateral biopsy is rarely practiced, and American guidelines recommend this procedure only if the contralateral testis is cryptorchid, shows marked atrophy, or suspicious ultrasonic changes, but not microlithiasis alone (124, 125). Our recommendation is to offer a contralateral biopsy to all patients at the time of surgery for the primary TGCT, perhaps with the exception of men older than 50, because of a minimal risk of GCNIS. Added benefits of a biopsy are a good assessment of the fertility potential of the other testicle, and the peace of mind for the patient concerning the possibility of bilateral cancer (126). Mild post-biopsy complications (3%) are rare and the procedure is well accepted by the patients (127). In young men with normal size testicles, two-site biopsies may be considered to decrease a chance of a false-negative biopsy (126). False-negative cases are relatively rare (128) but the procedure does not completely eliminate the risk of the second TGCT. In the Danish nationwide study, the cumulative incidence of the metachronous TGCT was 1.9% (after median 20-year follow-up) compared to 3.1% in a non-biopsied earlier cohort of patients (121). However, in a sub-cohort of Danish patients, in whom the contralateral biopsy was performed with a quality control and obligatory immunohistochemistry with GCNIS markers, the cumulative rate of the second cancer (after similar follow-up period rate) decreased to 0.95% (121).  

 

Technical aspects of surgical biopsy are important. The tissue fragment has to be sufficiently large (approx. 3x3x3 mm) and care has to be taken to avoid damage to the specimen, which should be dropped directly to a container with a fixative solution. For morphology evaluation Bouin's or similar solutions are preferred because formalin causes shrinkage artefacts, while for immunohistochemistry buffered formalin is preferred. As for any diagnostic testis biopsy, the contralateral biopsy evaluation must include immunohistochemical staining for at least one (better two) of the known markers for GCNIS, e.g., placental-like alkaline phosphatase (PLAP), OCT-3/4 (POU5F1), PDPN/M2A/D2-40, or AP2γ-TFP2AC (120, 122, 129). An example of immunohistochemical PLAP staining for GCNIS detection is shown in Figure 1.

SEMEN ANALYSIS

 

Poor spermatogenesis is a good indication that the patient may be at risk of harboring GCNIS (5, 68, 69, 75, 76, 130, 131). Semen analysis cannot as yet be used alone for detection of early stages of testicular cancer. However, it has been known for a long time that GCNIS cells may be occasionally found in semen (132). Over the years, progress has been made towards establishing better quality immunocytochemical detection of GCNIS or early invasive intratubular tumor cells in semen, and an improved automated double-staining assay has been developed (133, 134, 135). Unfortunately, the sensitivity of the cytological method is not high enough for screening, but it can be used as an additional part of semen analysis in experienced andrological centers performing immunohistochemistry (135). It is also not possible to use a micro-RNA (miRNA)-based assay, especially miRNA-371a-3p, a very promising serum marker for invasive TGCT, for detection of GCNIS in seminal fluid, despite that this embryonic-type miRNA is secreted by GCNIS cells (37). Even though some patients with GCNIS can have measurable miR-371a-3p in serum (136), the presence of the same miRNAs in normal germ cells in control patients with non-malignant conditions precludes using this assay in seminal fluid (137, 138, 139).

 

SERUM TUMOR MARKERS, DIAGNOSIS AND MONITORING OF OVERT TGCT

In the vast majority of cases a scrotal mass is usually the first presentation of testicular cancer, with tenderness reported by very few patients. In a few percent of testicular cancer cases, the presenting symptoms are the result of metastatic disease. They are usually uncharacteristic and may include lumbar pain, palpable abdominal mass, supra-clavicular lymph node enlargement, and in rare cases pulmonary symptoms (1).

 

The majority of primary and metastatic germ cell tumors secrete proteins and other biochemical products that can be detected in circulating blood. These biochemical serum tumor markers are very helpful in diagnosis and monitoring of testicular cancer (38, 124, 140).

 

The most important serum markers established in clinical practice are human chorionic gonadotropin (HCG), alpha-fetoprotein (AFP), and lactate dehydrogenase (LDH), the first two mainly useful to detect nonseminomas (1, 140). Among nonseminomatous tumors, choriocarcinoma, which resembles a gestational trophoblast, produces large quantities of HCG, while yolk sac tumor, which is similar in morphology to the embryonic yolk sac, secretes AFP (38, 140, 141). In addition, LDH may be secreted by both seminomas and nonseminomas. LDH levels in serum tend to be higher in patients harboring tumors with an increased copy number of chromosome 12p, consistent with the genomic location of the LDHB gene (142). Increased concentrations of LDH in the absence of AFP and HCG suggests the presence of seminoma. It is important to keep in mind that TGCT rarely occur in pure histological forms, and a subtype of seminoma with syncytiotrophoblastic cells can secrete HCG, which can be measurable in patients’ blood (14, 143). In preinvasive GCNIS and in most cases of pure seminoma, none of the above-mentioned markers are detectable in serum.

 

The most promising currently emerging serum markers are micro-RNAs (miRNA), among which miRNA 371a-3p is the best studied and most robust marker, and has already been proven valuable for detection of seminomas and nonseminomas, except for differentiated pure teratomas, in children and adults (38, 144, 145). Several large clinical studies clearly demonstrated that miRNA 371a-3p test has a much better specificity than the classical serum markers, and is especially useful for detection and monitoring of HCG- and AFP-negative seminomas (146, 147).

 

After diagnosis, careful clinical staging is necessary in each patient to decide for the most appropriate treatment strategy, and the measurements of circulating tumor markers are an important part of this process (1, 123, 124). There is a tendency for higher levels of tumor markers to be associated with a poorer prognosis. For example, the presence of syncytiotrophoblastic cells in the subtype of seminoma can lead to a mild increase of serum HCG, but the tumor responds well to treatment regardless of the presence of these cells, so more aggressive management should be avoided.

 

In addition to serum markers, scrotal ultrasonography is the first line diagnostic procedure. In general, seminoma is a homogenous tumor, whereas nonseminomas usually display heterogeneous patterns, with frequent hyperechoic calcified areas (148, 149). Additional imaging procedures help to evaluate the spread of disease, including CT scan of the chest and abdomen, or MRI (148, 149, 150).

 

Histopathological evaluation of the orchiectomy specimen is an essential part of the clinical staging. The important risk factors are tumor size, vascular/lymphatic invasion or the presence of tumor cells in rete testis, and the presence of pure embryonal carcinoma (152, 153).

 

The most commonly used staging and prognostic classification system is the TNM (tumor, node, metastases) System of the International Germ Cell Cancer Collaborative Group (GCCCG) and the American Joint Committee on Cancer (AJCC) (154). The stage grouping updated by the WHO Consensus (2), is shown in Table 2.

 

Table 2. Stage Grouping According to TNM Classification

Stage 0 (pTis): Germ cell neoplasia in situ

Stage IA (pT1, N0, M0, S0): Tumor limited to testis and epididymis

Stage IB (pT2-4): as IA but with vascular/lymphatic, tunica or scrotal invasion

Stage II (any pT, N1-3, M0, S0-1): Metastasis in lymph nodes, serum markers normal or moderately increased

      IIA (N1): lymph nodes <2 cm

     IIB (N2, S1): lymph nodes >2 cm but <5 cm

     IIC (N3, S1): lymph nodes >5 cm

Stage III (any pT, any N, M1, S0-3): Distant metastasis (spread beyond regional nodes)

     IIIA (M1a, S0-1): Spread to non-regional nodes or lung

     IIIB (M1a, S2): as IIIA but high serum markers

     IIIC (M1a-b, S3): distant metastasis to sites other than IIIA, or very high serum markers

Footnote: pT=primary tumor, N=regional lymph nodes, M=distant metastasis, S=serum tumor markers (X=unknown, not available)

 

Management of GCNIS and Testicular Germ Cell Cancer  

Early diagnosis of testicular neoplasia at the stage of GCNIS, followed by adequate treatment of GCNIS is capable of preventing progression to invasive tumors. Unfortunately, the vast majority of cases progress unnoticed to overt tumors. However, germ cell tumors are extremely radio- and chemo-sensitive and have apparently very high propensity to apoptosis, likely mediated by p53 (155, 156). Because of this sensitivity, and thanks to cisplatin-based chemotherapy regimens, TGCT is a highly curable malignancy, with more than 90% of patients reaching a sustained complete remission (157).

 

We give here only very general information concerning management of testicular cancer. The reader should consult specialized oncologic and urologic literature for management options pertinent to treatment-resistant tumors and metastatic disease.

 

TREATMENT OF GCNIS

 

Spontaneous regression of GCNIS has never been described and it is anticipated that all cases of GCNIS will eventually develop into TCGT. The following management options for GCNIS are available depending on a specific situation (158):

 

  • Orchiectomy - is the curative treatment with the highest assured success rate. It should always be performed on a testis with GCNIS or localized tumor when the second testis is not affected by malignancy (including GCNIS).
  • Radiotherapy - low dose radiotherapy is a good alternative to orchiectomy when GCNIS is present bilaterally or in the contralateral testis, so the patient can be spared a bilateral castration and lifelong androgen replacement therapy. The efficacy of radiotherapy with doses as low as 16 Gy was demonstrated in the early studies (159). Even though the lower dose better preserves the function of Leydig cells (160), more recent studies and current EAU guidelines recommend a dose of 18 Gy, given in fractions of 2Gy (123, 161). This dose of radiation will almost always destroy normal germ cells, so radiotherapy may be delayed in patients who wish to secure natural conception of a child.
  • Chemotherapy - is not an option to treat GCNIS, because a persistence or relapse has been reported in a high proportion of patients (121, 161). Furthermore, in some cases of extragonadal germ cell tumors treated with chemotherapy, testicular GCNIS progressed to metachronous overt testicular tumors (162). However, in patients with disseminated disease receiving chemotherapy, the risk of metachronous bilateral TGCT is lower (73, 163, 164).
  • Surveillance - is potentially hazardous since it increases the risk of metachronous bilateral TGCT (164) and GCNIS may progress to invasive cancer at any time. However, watchful surveillance may be an option after careful informed discussion of risks and monitoring with ultrasound examinations, especially if the patient wishes to defer treatment temporarily for the purpose of paternity.

 

In men who desire fatherhood, and in all very young men, cryopreservation of semen samples should be offered, if viable spermatozoa are detected.

 

TREATMENT OF OVERT SEMINOMAS AND NONSEMINOMAS

 

As far as the treatment of invasive germ cell tumors is concerned, the reader should consult the specialized urology and oncology guidelines.

 

Prior to surgery, all patients with TGCT should be offered full andrological evaluation and cryopreservation of semen. Endocrinologic evaluation of the patient should also preferably be done before surgery, with the reproductive hormone profile, including serum testosterone, gonadotropins, and inhibin B, if possible. This can be done together with the above-mentioned measurements of serum tumor markers. Pre-operative semen analysis and cryopreservation is especially important in patients with bilateral tumors or only one testicle. In the patients who have azoospermia or cryptozoospermia, and in whom it was impossible to retrieve sperm pre-operatively, testicular sperm extraction (TESE) at the time of orchiectomy (‘onco-TESE’) may be attempted in specialized centers (165). The best predicting factor of sperm retrieval is a small size of the tumor, because a chance of finding tubules with ongoing spermatogenesis increases with the distance from the tumor (166).

 

Radical orchiectomy remains the primary surgical treatment method of choice. Primary dissection of retroperitoneal lymph nodes (RPLND) was previously commonly practiced, especially in North America. The current consensus is to perform RPLND in experienced high-volume centers, and in selected cases, mainly in patients with nonseminoma and intermediate prognosis (1, 167, 168, 169). Nerve-sparing technique is preferred in order to avoid the loss of antegrade ejaculation, which is the commonest long-term complication of RPLND (169). 

 

Methods of post-surgical management of overt TGCT are variable, depending upon the histological type of tumor (seminoma vs nonseminoma), levels of serum markers and stage of disease and the presence of residual retroperitoneal masses (1, 115, 151, 167). Adjuvant radiotherapy, e.g., of the retroperitoneal/para-aortic field, which was previously routinely used, is no longer recommended, because of the long-term risk of secondary malignancies (1, 167, 168). 

 

Pure seminomas have a good prognosis and around 80% of patients with stage I seminoma (tumor confined to the testis) do not require any treatment after the surgery, thus most of the centers practice a surveillance strategy (151, 168, 170). Nonseminomas have a somewhat poorer prognosis (relapse rate is around 30%) and in some centers, patients with nonseminoma and high risk of relapse are treated with one course of adjuvant chemotherapy with cisplatin and etoposide (171, 172), but most centers currently recommend a surveillance strategy (1, 151, 152, 168, 172).

 

The most common post-surgical management of disseminated disease is systemic chemotherapy with a combination of cytotoxic drugs. The standard first line chemotherapy regimen is BEP (bleomycin, etoposide and cisplatin), administered in 3 or 4 cycles, depending on the patient’s prognosis (173, 174). It is very important to make a dynamic assessment of the progress of treatment through the early stages of chemotherapy, thus monitoring of serum markers is obligatory. Post-chemotherapy retroperitoneal surgery should be performed in patients with a residual tumor after BEP (151). In patients with relapse after first line treatment, salvage regimens and complex surgery for residual tumors need to be performed. Overall, the management is difficult, thus it should be carried out in specialized tertiary centers (1, 167).

 

The majority of relapses occur within 2 years of the initial treatment but late relapses are observed in some cases, therefore individual management of each patient and lifetime follow-up is advocated in some patients (1, 151, 167). Andrological follow-up should be coordinated with the post-surgery oncological management and controls (see the section on ‘Endocrine problems and late effects’).

 

SEX CORD-STROMAL TUMORS OF THE TESTIS

 

In adults, sex cord-stromal tumors of the testis are found in less than 5% of all testicular tumors, whereas in children, these tumors are more common and account for approximately 25% of cases (123, 175, 176, 177, 178). Most of these tumors are benign, only around 5% have malignant characteristics (2). The classification of the tumors by WHO (2016) is shown in a simplified form in Table 3 (2).

 

Sex cord-stromal tumors are derived from testicular somatic cells; Leydig cells, and Sertoli/granulosa cells. Despite a different cell of origin, some stromal tumors are sometimes misinterpreted as seminoma. A number of features can be used to distinguish sex cord-stromal tumors from germ cell tumors; Inhibin A and B are the best serum markers for this purpose (179, 180, 181). Inhibin A appears to be a common immunohistochemical marker for sex cord-stromal tumors, including Leydig cell, Sertoli cell and juvenile granulosa cell tumors (179, 182). Sertoli cell tumors are positive for anti-Mullerian hormone (183) and GATA-4 (184), while Leydig cell tumors express steroidogenic enzymes and INSL3 (185). Other useful immunohistochemical markers, include SOX9, calretinin, CD99, SF1 (2).

 

Table 3. Sex Cord-Stromal Tumors of the Testis, Adapted from WHO Classification (ref. 2)

 

·                Leydig cell tumor (8650/1)

Malignant Leydig cell tumor (8650/3)

·                Sertoli cell tumor (SCT) (8640/1)
Malignant SCT (8640/3)
Large cell calcifying SCT (8642/1)
Intratubular large cell calcifying SCT (8643/1)

·                Granulosa cell tumors

Juvenile-type granulosa cell tumor (8622/1)

Adult-type granulosa cell tumor (8620/1)

·                The fibroma-thecoma tumors (8600/0)

·                Mixed sex cord-stromal tumors (8592/1)

·                Unclassified sex cord-stromal tumors (8591/1)

*Footnote to Table 2: The codes in parentheses are from the International Classification of Diseases for Oncology (ICD-O-3).

 

Leydig Cell Hyperplasia and Tumors



Leydig cells are located in the interstitial compartment of the testis and are involved in the development of secondary male characteristics and maintenance of spermatogenesis by secretion of testosterone. Although Leydig cells in adult men are considered to be a terminally differentiated and mitotically quiescent cell type, in various disorders of testicular function, focal or diffuse Leydig cell hyperplasia is very common.  Micronodules of Leydig cells are frequently seen in certain conditions associated with severe decrease of spermatogenesis or germinal aplasia, such as the Sertoli-cell-only syndrome (Del Castillo syndrome), cryptorchidism, or Klinefelter syndrome, when the nodules can be particularly florid (186, 187). A term 'Leydig cell adenoma' is used when the size of a nodule exceeds several- fold the diameter of a seminiferous tubule. It is unknown whether Leydig cell adenomas can progress further to form overt Leydig cell tumors, but even if it were the case, it is exceedingly rare. Morphological heterogeneity of hyperplastic Leydig cells is noticeable in some cases, and it has been shown that the micronodules contain a large proportion of immature Leydig cells (186, 187).

 

The mechanism of Leydig cell hyperplasia and larger nodules in the human male is still poorly understood. Leydig cell nodules of variable-size are common in patients with TDS and infertility, and the functional insufficiency of Leydig cells is often reflected by decreased testosterone/LH ratio whereas patients with overt Leydig cell tumors usually have an increased testosterone/LH ratio and high estradiol (186, 188). The disruption of hypothalamo-pituitary-testicular axis leading to an excessive stimulation of Leydig cells by LH can play a central role (186). This in turn leads to an increased renewal of immature, adult-type Leydig cells from their precursors. The immature cells are characterized by low numbers of Reinke crystals, a relatively high expression of a mesenchymal factor DLK1 and low amounts of INSL3 (187, 189, 190). However, Leydig cell hyperplasia is distinct from tumors that are usually solitary. Leydig cell hyperplasia and adenomas can be easily induced in rodents by administration of estrogens, gonadotropins, and a wide range of chemical compounds. Whether or not humans would be similarly susceptible to environmental effects remains to be elucidated.

 

Leydig cell tumors account for up to 5% of testicular neoplasms and occur in all age groups (2, 178, 191). Approximately 20% are found in boys, most often between five and ten years of age. There is perception that the prevalence of benign Leydig cell tumors among the adults is greater than previously thought, possibly thanks to improved detection of small nonpalpable nodules by modern imaging methods. Higher incidence numbers have also been reported in fertility centers that actively scan the testes of infertile patients (192).

 

In a subset of cases of Leydig cell tumors, activating mutations of the LH receptor (193, 194, 195) or G proteins (196, 197) can be detected. Constitutively activating mutations of LH receptor cause early Leydig cell hyperplasia and precocious puberty (193, 195, 197). Similarly, constitutively activating mutations of Gs-protein in Leydig cells or inactivation of PKAR1A (protein kinase cyclic adenosine monophosphate-dependent regulatory type 1 alpha) lead to hyperplasia and endocrine hyperactivity (178, 197). In adult Leydig cell tumors, germline fumarate hydratase mutations have been identified in a few cases which also had hereditary leiomyomatosis and renal cell cancer (199). 

 

Precocious puberty (so-called testotoxicosis) is the presenting symptom in most of cases of Leydig cell adenomas or tumors in children, due to the excessive androgen production, mainly testosterone that causes growth of penis, pubic hair, accelerated skeletal and muscle growth, advancement of bone age, skin changes (acne, comedos, hair greasing), and adult-type odor of sweat. Androgen secretion in the pediatric cases is gonadotropin independent, and therefore LH and FSH remain low in spite of external signs of puberty (195). Approximately 10% of the boys also have gynecomastia that is caused by estrogens produced in excess due to aromatase activity in some of the tumors or peripheral aromatization of testosterone. It is important to keep in mind that transient gynecomastia or pseudo-gynecomastia can occur in newborns, in obese boys, and at puberty as a non-pathological condition (200). In adults, gynecomastia is a frequent condition with a reported prevalence of 32–65%, depending on the age and the criteria used for definition (summarized in 201). Gynecomastia is sometimes associated with loss of libido, impotence, and infertility (201). However, Leydig cell tumor is the cause in only 1-2% of cases, and the excessive androgen secretion by these tumors rarely causes notable effects in adults (202). Malignant tumors are hormonally active only in exceptional cases (2, 178).

 

In children, Leydig cell tumors are always benign and can be treated with surgical enucleation when the tumor is encapsulated (203). In adults malignant Leydig cell tumors have been found in 10-15 % of patients, and inguinal orchiectomy is often used (204), while testis-sparing enucleation remains an option, if there are no signs of malignancy in the frozen preparation (205). The presence of cytologic atypia, necrosis, angiolymphatic invasion, increased mitotic activity, atypical mitotic figures, infiltrative margins, extension beyond testicular parenchyma, and DNA aneuploidy are associated with metastatic behavior in Leydig cell tumors (178, 202, 206) (see Figure 5).

Figure 5. Histology of a Leydig cell tumor.  The appearance of tumor cells resembles normal Leydig cells. A section stained with hematoxylin-eosin (HE).

 

Malignant Leydig cell tumors do not respond favorably to conventional chemotherapy and irradiation (204). Survival time has ranged from 2 months to 17 years (median, 2 years), and metastases have been detected as late as nine years after the diagnosis (178, 202). Metastatic Leydig cell tumors require salvage surgery, radiotherapy and intensive chemotherapy regimens but in most cases the outcome is poor (188, 207). Therefore, follow-up of patients with malignant Leydig cell tumors has to be life-long. The remaining testis may be irreversibly damaged by longstanding high estrogen levels, resulting in both permanent infertility and hypoandrogenism (202, 206).

 

Testicular Adrenal Rest Tumors (TART)

 

Excessive secretion of adrenocorticotropin (ACTH) in 21-hydroxylase deficiency (congenital adrenal hyperplasia, CAH) or Nelson syndrome (post adrenalectomy status) may lead to development of hyperplastic interstitial nodules called testicular adrenal rests tumors (TART) resembling Leydig cell tumor or hyperplasia (191, 208, 209). These cells are hormonally active in secreting androgens. It is important to remember that the adrenal rests are almost invariably bilateral, whereas the Leydig cell tumors are usually unilateral. Adrenal rests can be treated by appropriate glucocorticoid substitution of the patient, which leads to gradual regression of the 'tumor' in 75 percent of cases (208, 209). TART have to be distinguished from benign hyperplasia, adenomas and malignant Leydig cell tumors (185).

 

Histopathological and endocrine evaluation covering both testicular and adrenal steroids and pituitary gonadotropins and ACTH are important in the differential diagnosis (185, 208). It would be an error to orchidectomize the CAH patients with TART, since the tumors are always benign and only some of them continue to be active after appropriate glucocorticoid substitution. In the patients who do not respond well to glucocorticoid replacement, or develop fibrosis, testis sparing surgery with enucleation of the larger nodules can be considered, if the testicles have become so large that they cause discomfort for the patient (209).

 

Sertoli Cell Tumors



Sertoli cells are the somatic cells in the seminiferous epithelium giving structural, metabolic, and hormonal support to spermatogenic cells. Sertoli cells cease their proliferation at puberty. In rare infantile cases, multiple foci of proliferating Sertoli cells have been described and proposed to be early intratubular forms of Sertoli cell tumors (210). The classification of these tumors according to the WHO (2, 178), is shown in Table 3.

 

Sertoli cell tumors often occur as a part of multiple neoplasia syndromes (see below). Rare Sertoli cell tumors which do not belong to a syndrome are called ‘not otherwise specified’ (NOS). About 5% of these tumors are malignant and can metastasize. The age of patients ranges from 18 to 80 years, but most of them are young adults (median age 30). Out of 60 patients, only four were younger than 20 years old in the series reviewed by Young et al. (211). The tumors typically are composed of sex cord cells with tubular differentiation, with a subset of tumors hyalinized, previously classified as sclerosing variant (2).  Some tumors often contain lipid droplets but do not show any endocrine activity. The molecular origin is known only in a small proportion of tumors with sclerosing appearance, in which CTNNB1mutations causing nuclear accumulation of β-catenin were identified (212). The tumors occurred in descended testes and were always unilateral. An infiltrative margin was found in four cases, but most of the tumors were well demarcated. The tumors were hormonally inactive, and only two patients with alcoholic cirrhosis also had gynecomastia. Eighteen pediatric cases were reported from the Kiel Pediatric Tumor Registry (176), but perhaps the histopathologic pattern was somewhat different, because the age of the children was very young, ranging from 0 to 14 months (median 4 months). Juvenile Sertoli cell tumors often showed infiltrative growth into adjacent tissue, dense cellularity and considerable proliferative activity. However, after surgical excision no local recurrences and no metastases occurred. Thus, these patients have a good prognosis. These Sertoli cell tumors can be treated by orchiectomy, and retroperitoneal lymphadenectomy is indicated only when there is radiographically detected retroperitoneal involvement (213).

 

Calcifying Sertoli cell tumors, are frequently found in association with two distinct multiple neoplasm syndromes, Carney complex and Peutz-Jeghers syndrome, however in the latter syndrome, the tumors belong to a distinct ‘intratubular’ morphological group (2, 178, 214). Association of large-cell calcifying Sertoli cell tumors with other neoplasms, particularly heart myxomas in Carney complex and gastrointestinal tumors in Peutz-Jeghers syndrome, should be kept in mind to reach an early diagnosis of these potentially fatal diseases.

 

The Carney complex is characterized by skin myxomas, heart myxomas, skin pigmentations, adrenal and testicular tumors, but other tumors can also occur (215). The testicular tumors are large-cell calcifying Sertoli cell tumors that are multifocal and bilateral, and should be distinguished from teratomas (Figure 6) (178, 216, 217). The tumors appear usually during the second decade of life (218). Only one malignant case has been reported in association with Carney complex (in an adult patient), whereas seven malignant tumors were reported in other patients with large-cell calcifying Sertoli cell tumors (218). These patients were older than 25 years. The malignant cases were unilateral and solitary in contrast to bilateral and multifocal occurrence of testicular tumors in Carney complex. Large-cell calcifying Sertoli cell tumors are usually not hormonally active, although elevated levels of serum inhibin B or testosterone have been reported, but other tumors of Carney complex, including Leydig cell tumors, can cause endocrine manifestations (181, 213, 214).

Figure 6. Large cell calcifying Sertoli cell tumor isolated from a 12-year-old boy. The neoplastic tubules contain only large pale Sertoli cells and visible calcifications in the lumen (stained with PAS). Adjacent normal tubules show advanced spermatogenesis.

 

Two genetic loci for Carney complex have been identified on chromosome 2p16 (196) and 17q23-24 (219, 220). Germline mutations identified in Carney complex most often occur in type I-alpha regulatory subunit of protein kinase A, PRKAR1A (221, 222). Inactivating mutations of phosphodiesterase 11A212 and phosphodiesterase 8B213 are associated with bilateral adrenocortical hyperplasia in Carney complex patients. Genetic variation of the phosphodiesterase 11A gene can modify the development of the testicular tumors (223). Molecular genetic diagnosis is available to many of these patients. However, only a part of the mutations occur in the germ line, and therefore genetic analysis should be performed on affected tissues, such as the tumors, in cases of somatic cell line mutations.

 

In Peutz-Jeghers syndrome Sertoli cell tumors are similar in appearance to those found in Carney complex patients, but can be distinguished by typical intratubular proliferation of lightly eosinophilic cells with prominent basement membrane deposits, and occasionally, features of ovarian sex-cord tumors with annular tubules (178). Thus, the tumors are described as intratubular large-cell hyalinizing Sertoli cell neoplasia. These tumors may have strong aromatase activity and therefore be associated with gynecomastia and advanced bone age (224). No malignant testicular tumors have been reported in Peutz-Jeghers patients, but they have a highly increased risk of other neoplasms, especially colorectal, breast, pancreatic and ovarian cancers (214). Germline loss-of-function mutations in the STK11/LKB1 gene that encodes for a serine-threonine kinase causes Peutz-Jeghers syndrome in the majority of patients, allowing molecular genetic diagnostics (214, 224, 225).

 

Patients with the Sertoli cell tumors should be treated conservatively during childhood to give them a possibility for sperm banking before orchiectomy comes necessary (226).  Autosomal dominant inheritance should be considered in genetic counselling. The patients should be frequently controlled with ultrasound examinations to follow changes in the tumors size, and reproductive hormone measurements. If precocious puberty and/or gynecomastia appear, aromatase inhibitors and antiandrogens can be used to prevent estrogen formation and androgen action (219, 226). There are currently no clear guidelines for the treatment of metastatic Sertoli cell cancers, but orchiectomy and retroperitoneal lymph node dissection are usually performed combined with individualized chemotherapy and radiotherapy (227). Without surgery, the metastatic disease progresses and 20-month median survival time of patients was reported (IQR: 6–30 months) (227).

 

 Granulosa Cell Tumors



Juvenile-type granulosa cell tumors are rare but are the most common somatic testicular tumors in infants and occur during the first 6 months after birth (2, 176, 178, 228). These tumors were described in patients with undescended testes with abnormal sex chromosomes and ambiguous genitalia (176). Prominent differentiation into follicles and immature nuclei distinguishes juvenile granulosa cell tumors from other Sertoli cell tumors that express tubular differentiation (178, 229). Most of the immunohistochemical markers in these tumor types are similar, e.g., inhibin, calretinin, but also distinct, e.g., expression of FOXL2 (229). Juvenile granulosa cell tumors always have a good prognosis (230). Testicular tumors do not show endocrine hyperactivity, in contrast to ovarian juvenile granulosa cell tumors. Aberrant WNT signaling (231) and stimulatory G-protein mediated signaling (232) have been linked with juvenile granulosa cell tumors. The patients should be managed comprehensively by a multidisciplinary DSD team, and the treatment of tumors may require orchiectomy.

 

Adult-type granulosa cell tumors are comparable to the ovarian tumors, but are extremely rare (2, 178, 233). These tumors occur in adults at an average age of 42 years. Twenty percent of the patients have shown gynecomastia due to the hormonal activity of the tumor. Most of the tumors are benign, but some malignant cases have also been reported (230). Mutations in FOXL2 have been reported in adult ovarian granulosa cell tumors but only few secondary mutations have been found in testicular granulosa cell tumors (234, 235). The treatment is primarily surgical.

 

Fibroma-Thecoma Tumors

These exceedingly rare neoplasms are composed of fibroblastic cells of testicular stroma or tunica albuginea (2). These tumors are reported to be benign.

 

Mixed and Unclassified Sex Cord-Stromal Tumors

Tumors consisting of more than one stromal or tubular component or have indeterminate morphology are classified as mixed and unclassified sex cord-stromal tumors (2). Sex cord-stromal tumors can contain combinations of Leydig, Sertoli, granulosa, and theca cells, and are therefore called mixed tumors (178). Leydig cells can be difficult to recognize in these tumors. These tumors are rare and can occur at any age. Depending on the predominant cell type the tumors may behave differently. Gynecomastia, as a sign of endocrine activity, can be found in 10% of patients (2). These tumors are always benign in children, but in adults, malignancy can be found (232). Thus, most of the patients can be treated by orchiectomy, and lymph node dissection is indicated only in cases with overt malignant features on microscopic examination.

 

OTHER TESTICULAR TUMORS

 

Other tumors occurring in the testis are divided into miscellaneous and hematolymphoid tumors (2, 178). The first group includes ovarian epithelial-type tumors, serous or mucinous cystadenomas, adenocarcinomas, Brenner tumor, xantogranuloma, and hemangioma. The hematolymphoid tumors comprise malignant lymphomas (B-cell, NK/T-cell or follicular), plasmacytoma, myeloid sarcoma and Rosai-Dorfman disease (2, 178). In addition, testicular spread of malignant acute leukemia is common in young boys, and metastases from other solid tumors including the prostate gland, colon, kidney, stomach, pancreas, and malignant melanoma can be found in the testis of adults.

 

ENDOCRINE PROBLEMS AND LATE EFFECTS IN TESTICULAR CANCER PATIENTS

 

Testis Dysfunction and Fertility Issues

 

Relative imbalance of androgen signaling (excess or deficiency) causes the most pronounced secondary endocrine symptoms associated with testicular tumors. Testosterone is produced by tumors, such as Leydig cell tumors, or by normal Leydig cells stimulated by large amounts of hCG from some germ cell tumors. Excess of androgens would lead to precocious puberty in children (193, 194, 195). In addition, aromatization of androgens leads to a relative excess of estrogens, which causes impairment of spermatogenesis in adults and gynecomastia at any age (224, 236).

 

Testicular dysfunction in young adult patients with testicular cancer, who usually are in their best reproductive age, is a serious clinical problem. Patients with TGCT have poor spermatogenesis and decreased fertility even before the overt tumor has developed (67, 68, 69, 130) and before cytotoxic treatment (237, 238). Pathological features can include oligozoospermia or azoospermia, moderately decreased testosterone and elevated LH levels. If testicular biopsies are taken, a variable degree of testicular dysgenesis or atrophy is often seen, and in some cases further complicated by the presence of GCNIS. Examination of a contralateral biopsy in a patient with a unilateral tumor may show a similar picture; with at least 5-7% risk of the presence of GCNIS cells (71, 126).

 

Testicular function is further disturbed by treatment of malignancy. In recent years there is growing concern about adverse late effects of irradiation and chemotherapy, which induce severe impairment of spermatogenesis and DNA damage (1, 167, 168, 239, 240).  Refinement of the dosage must be considered in each patient individually, to eradicate the neoplasm with least possible damage to the endocrine function. The eradication of GCNIS or a tumor by irradiation in bilateral cancer cases leads also invariably to the disappearance of all germ cells and sterility. This underlines the importance of semen cryopreservation before treatment, which will allow assisted reproduction treatment, if needed (238, 241).

 

All patients treated for testicular cancer require assessment of their reproductive hormones and spermatogenic capacity by semen analysis with respect to their future fertility. If semen banking or pre-operative sperm retrieval is not possible or if the patient has azoospermia or cryptozoospermia, testicular sperm extraction (TESE) at the time of orchiectomy (‘onco-TESE’) may be the only chance of fertility, and can be attempted in specialized centers (165, 166). TESE and subsequent intracytoplasmic sperm injection (ICSI) may be also an option for fertility treatment in some cases of post-chemotherapy azoospermia (242).

 

Fertility preservation is a serious challenge in children and young adolescents. In the adolescent boys who are unable to ejaculate, penile vibratory stimulation or electroejaculation can be considered (243). However, there is currently no treatment for prepubertal boys. There are ongoing studies attempting to optimize protocols of cryopreservation of immature testis tissue before cytotoxic treatment or orchiectomy (244). This approach would require in vitro or in vivoinduction of spermatogenesis and gametogenesis, which has not yet been achieved satisfactorily in humans, but studies in experimental animals, including nonhuman primates are promising (245).

 

Long-Term Sequelae and Quality of Life

 

All patients treated for testicular cancer require monitoring of their reproductive hormone profiles not only with respect to their fertility, if paternity is desired, but also risk of testosterone deficiency and ensuing symptoms (246, 247, 248).

 

An increased risk of cardiovascular disease is present in TGCT survivors who have been treated with radio- or chemotherapy (249, 250) but a recent Danish study found that the risk decreases during long-term follow-up, although it remains elevated (251). Additional problems in survivors of TGCT treated with radiotherapy or chemotherapy are increased risk of peripheral neuropathy or ototoxicity (after chemotherapy) and sexual dysfunction (after chemotherapy and radiotherapy) (239, 240, 250, 251, 252). 

 

Survivors of metastatic TGCT have increased mortality rates when compared to the general male population. A study from Norway reported that most of the excess deaths occurred within a decade from diagnosis due to the testicular cancer, but during a longer follow-up, additional excess deaths appeared in patients treated with radiotherapy or cisplatin-based chemotherapy, mainly due to non-TGCT second cancers; primarily gastric, pancreatic or bladder tumors (253, 254). A population-based large study from the US found an increased risk of leukemia after chemotherapy and a marked excess of solid tumors in patients treated with radiotherapy, which can appear after long follow-up (255). This study confirmed previously published worrying reports from several centers, which called for reducing the use of radiotherapy and radiologic imaging procedures during the follow-up (256).

 

Of importance for the testicular cancer survivors are also quality of life issues related to prolonged anxiety, depression and psychological stress, and loss in socioeconomic status or unemployment, leading to excess of suicide among the patients (167, 252, 254, 257, 258, 255, 259, 260). Hence, a psychologist/social advisor should be added to the multidisciplinary team of experts caring for survivors of TGCT. Clearly, the focus of the current comprehensive care has moved from survival to survivorship of the patients after their recovery from testicular cancer.

 

CONCLUSION

 

Key take home points are shown in figure 7.

Figure 7. Key take home points.

 

ACKNOWLEDGEMENTS  

 

The authors thank Prof. Niels E. Skakkebæk for his mentorship, and the research and clinical teams at their departments for the contribution to the studies summarized in this review. The work was supported by grants from numerous foundations, with the biggest contributions from the Danish Cancer Society, the Lundbeck Foundation, the Svend Andersen Foundation, the Danish Advanced Technology Foundation, Sigrid Juselius Foundation and Novo Nordisk Foundation.

 

REFERENCES

 

  1. Rajpert-De Meyts E, McGlynn KA, Okamoto K, Jewett MAS, Bokemeyer C. Testicular Germ Cell Tumours. Lancet 2016; 387 (10029): 1762-1774.org/10.1016/S0140-6736(15)00991-5
  2. Moch H, Humphrey PA, Ulbright TM, Reuter VE, (Eds). WHO Classification of Tumours of the Urinary System and Male Genital Organs, 4th Edition.IARC Press, Lyon 2016.
  3. Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular Dysgenesis Syndrome: An increasingly common developmental disorder with environmental aspects. Hum Reprod 16: 972-978, 2001.
  4. Rajpert-De Meyts E. Developmental model for the pathogenesis of testicular carcinoma in situ: Environmental and genetic aspects. Hum Reprod Update 12: 303-323, 2006.
  5. Skakkebæk NE, Rajpert-De Meyts E, Buck Louis GM, Toppari J, Andersson AM, Eisenberg ML, Jensen TK, Jørgensen N, Swan SH, Sapra KJ, Ziebe S, Priskorn L, Juul A. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol Rev 96: 55-97, 2016.
  6. Skotheim RI, Lind GE, Monni O, Nesland JM, Abeler VM, Fosså SD, Duale N, Brunborg G, Kallioniemi O, Andrews PW, Lothe RA. Differentiation of human embryonal carcinomas in vitro and in vivo reveals expression profiles relevant to normal development. Cancer Res 65(13): 5588-98, 2005.
  7. Solter D. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nature Rev Genetics 7: 319-327, 2006.
  8. Oosterhuis JW, Looijenga LHJ. Human germ cell tumours from a developmental perspective. Nat Rev Cancer 19:522–537, 2019.
  9. Busch J, Seidel C, Zengerling F. Male Extragonadal Germ Cell Tumors of the Adult. Review. Oncol Res Treat. 39(3):140-4, 2016. doi: 10.1159/000444271.
  10. Aguirre D, Nieto K, Lazos M, Peña YR, Palma I, Kofman-Alfaro S, Queipo G. Extragonadal germ cell tumors are often associated with Klinefelter syndrome. Hum Pathol 37: 477-480, 2006.
  11. Fosså SD, Aass N, Heilo A, Daugaard G, Skakkebaek NE, Stenwig AE, Nesland JM, Looijenga LH, Oosterhuis JW. Testicular carcinoma in situ in patients with extragonadal germ-cell tumours: the clinical role of pretreatment biopsy. Ann Oncol. 14(9):1412-1418, 2003.
  12. Scholz M, Zehender M, Thalmann GN, Borner M, Thöni H, Studer UE. Extragonadal retroperitoneal germ cell tumor: evidence of origin in the testis. Ann Oncol. 2002; 13(1):121-124.
  13. Berney DM, Looijenga LH, Idrees M, Oosterhuis JW, Rajpert-De Meyts E, Ulbright TM, Skakkebaek NE. Germ cell neoplasia in situ (GCNIS): evolution of the current nomenclature for testicular pre-invasive germ cell malignancy. Histopathology.; 69(1):7-10, 2016. doi: 10.1111/his.12958.
  14. Berney DM, Cree I, Rao V, Moch H, Srigley JR, Tsuzuki T, Amin MB, Comperat EM, Hartmann A, Menon S, Netto GJ, Rubin MA, Turajlic S, Raspollini MR, Tickoo SK. An Introduction to the WHO 5th Edition 2022 Classification of Testicular tumours. Histopathology. 2022 May 3. doi: 10.1111/his.14675. Epub ahead of print. PMID: 35502823.
  15. Skakkebaek NE. Possible carcinoma-in-situ of the testis. Lancet ii: 516-517, 1972.
  16. Skakkebaek NE, Berthelsen JG, Giwercman A, Müller J. Carcinoma-in-situ of the testis. Possible origin from gonocytes and precursor of all types of germ cell tumours except spermatocytoma. Int J Androl 10: 19-28, 1987.
  17. Sonne SB, Almstrup K, Dalgaard M, Juncker AS, Edsgard D, Ruban L, Harrison NJ, Schwager C, Abdollahi A, Huber PE, Brunak S, Gjerdrum LM, Moore HD, Andrews PW, Skakkebaek NE, Rajpert-De Meyts E, Leffers H. Analysis of gene expression profiles of microdissected cell populations indicates that testicular carcinoma in situ is an arrested gonocyte. Cancer Res 69: 5241-5250, 2009.
  18. Jørgensen A, Lindhardt Johansen M, Juul A, Skakkebaek NE, Main KM, Rajpert-De Meyts E. Pathogenesis of germ cell neoplasia in testicular dysgenesis and disorders of sex development. Semin Cell Dev Biol 2015; 45: 124-137. dx.doi.org/10.1016/j.semcdb.2015.09.013.
  19. Cools M, Pleskacova J, Stoop H, Hoebeke P, Van Laecke E, Drop SL, Lebl J, Oosterhuis JW, Looijenga LH, Wolffenbuttel KP; Mosaicism Collaborative Group. Gonadal pathology and tumor risk in relation to clinical characteristics in patients with 45,X/46,XY mosaicism. J Clin Endocrinol Metab. 96: E1171-1180, 2011.
  20. Matsumoto F, Matsuyama S, Matsui F, Yazawa K, Matsuoka K. Variation of gonadal dysgenesis and tumor risk in patients with 45,X/46,XY mosaicism. Urology 137: 157-160, 2020.
  21. Jørgensen N, Müller J, Jaubert F, Clausen OP, Skakkebæk NE. Heterogeneity of gonadoblastoma germ cells: similarities with immature germ cells, spermatogonia and testicular carcinoma in situ cells. Histopathology 30,177-186, 1997.
  22. Hersmus R, Kalfa N, de Leeuw B, Stoop H, Oosterhuis JW, de Krijger R, Wolffenbuttel KP, Drop SL, Veitia RA, Fellous M, Jaubert F, Looijenga LH. FOXL2 and SOX9 as parameters of female and male gonadal differentiation in patients with various forms of disorders of sex development (DSD). J Pathol 215: 31-38, 2008.
  23. Guminska A, Oszukowska E, Kuzanski W, Sosnowski M, Wolski JK, Walczak-Jedrzejowska R, Marchlewska K, Niedzielski J, Kula K, Slowikowska-Hilczer J. Less advanced testicular dysgenesis is associated by a higher prevalence of germ cell neoplasia. Int J Androl 33: e153-e62, 2010.
  24. Chemes HE, Venara M, Del Rey G, Arcari AJ, Musse MP, Papazian R, Forclaz V, Gottlieb S. Is a CIS phenotype apparent in children with Disorders of Sex Development? Milder testicular dysgenesis is associated with a higher risk of malignancy. Andrology. 2015; 3(1): 59-69. doi: 10.1111/andr.301. PMID: 25598272.
  25. Jørgensen N, Rajpert-De Meyts E, Græm N, Müller J, Giwercman A, Skakkebaek NE. Expression of immunohistochemical markers for testicular carcinoma in situ by normal fetal human germ cells. Lab Invest 72: 223-231, 1995.
  26. Rajpert-De Meyts E, Nielsen JE, Skakkebæk NE, Almstrup K. Diagnostic markers for germ cell neoplasms: from placental-like alkaline phosphatase to micro-RNAs. Folia Histochem Cytobiol 2015; 53(3): 177-188. doi: 10.5603/FHC.a2015.0020.
  27. Almstrup K, Hoei-Hansen CE, Wirkner U, Blake J, Schwager C, Ansorge W, Nielsen JE, Skakkebaek NE, Rajpert-De Meyts E, Leffers H. Embryonic stem cell-like features of testicular carcinoma in situ revealed by genome-wide gene expression profiling. Cancer Res 64: 4736-4743, 2004.
  28. Alagaratnam S, Lind GE, Kraggerud SM, Lothe RA, Skotheim RI. The testicular germ cell tumour transcriptome. Int J Androl 34: e133-e150, 2011.
  29. Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, van Roozendaal KE, van Zoelen EJ, Weber RF, Wolffenbuttel KP, van Dekken H, Honecker F, Bokemeyer C, Perlman EJ, Schneider DT, Kononen J, Sauter G, Oosterhuis JW. POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res 63: 2244-2250, 2003.
  30. Hoei-Hansen CE, Nielsen JE, Almstrup K, Sonne SB, Graem N, Skakkebaek NE, Leffers H, Rajpert-De Meyts E. Transcription factor AP-2gamma is a developmentally regulated marker of testicular carcinoma in situ and germ cell tumors. Clin Cancer Res 10: 8521-8530, 2004.
  31. Hoei-Hansen CE, Almstrup K, Nielsen JE, Brask Sonne S, Graem N, Skakkebaek NE, Leffers H, Rajpert-De Meyts E. Stem cell pluripotency factor NANOG is expressed in human fetal gonocytes, testicular carcinoma in situ and germ cell tumours. Histopathology 47: 48-56, 2005.
  32. Hart AH, Hartley L, Parker K, Ibrahim M, Looijenga LH, Pauchnik M, Chow CW, Robb L. The pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer 104: 2092-2098, 2005.
  33. Rajpert-De Meyts E, Skakkebæk NE. Expression of the c-kit protein product in carcinoma in situ and invasive germ cell tumours. Int J Androl 17: 85-92, 1994.
  34. Bártková J, Bártek J, Lukás J, Vojtĕsek B, Stasková Z, Rejthar A, Kovarík J, Midgley CA, Lane DP. p53 protein alterations in human testicular cancer including pre-invasive intratubular germ-cell neoplasia. Int J Cancer 49: 196-202, 1991.
  35. West JA, Viswanathan SR, Yabuuchi A, Cunniff K, Takeuchi A, Park IH, Sero JE, Zhu H, Perez-Atayde A, Frazier AL, Surani MA, Daley GQ. A role for Lin28 in primordial germ-cell development and germ-cell malignancy. Nature 460: 909-913, 2009.
  36. Gillis AJ, Stoop H, Biermann K, van Gurp RJ, Swartzman E, Cribbes S, Ferlinz A, Shannon M, Oosterhuis JW, Looijenga LH. Expression and interdependencies of pluripotency factors LIN28, OCT3/4, NANOG and SOX2 in human testicular germ cells and tumours of the testis. Int J Androl 34: e160-e174, 2011.
  37. Novotny GW, Belling KC, Bramsen JB, Nielsen JE, Bork-Jensen J, Almstrup K, Sonne SB, Kjems J, Rajpert-De Meyts E, Leffers H. MicroRNA expression profiling of carcinoma in situ cells of the testis. Endocr Relat Cancer 19:365-379, 2012.
  38. Almstrup K, Lobo J, Mørup N, Belge G, Rajpert-De Meyts E, Looijenga LHJ, Dieckmann KP. Application of miRNAs in the diagnosis and monitoring of testicular germ cell tumours. Nat Rev Urol. 17: 201-213, 2020.
  39. Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, Skotheim RI, Abeler VM, Rajpert-De Meyts E, Lothe RA. Molecular characteristics of malignant ovarian germ cell tumours and comparison to testicular counterparts: Implications for pathogenesis. Endocrine Reviews 2013; 34: 339-376.
  40. Palmer RD, Barbosa-Morais NL, Gooding EL, Muralidhar B, Thornton CM, Pett MR, Roberts I, Schneider DT, Thorne N, Tavaré S, Nicholson JC, Coleman N; Children's Cancer and Leukaemia Group. Pediatric malignant germ cell tumors show characteristic transcriptome profiles. Cancer Res 68: 4239-4247, 2008.
  41. Palmer RD, Murray MJ, Saini HK, van Dongen S, Abreu-Goodger C, Muralidhar B, Pett MR, Thornton CM, Nicholson JC, Enright AJ, Coleman N; Children's Cancer and Leukaemia Group. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Res 70: 2911-2923, 2010.
  42. Manivel JC, Simonton S, Wold LE, Dehner LP. Absence of intratubular germ cell neoplasia in testicular yolk sac tumors in children. A histochemical and immunohistochemical study. Arch Pathol Lab Med 112: 641-645, 1988.
  43. Mosbech CH, Rechnitzer C, Brok JS, Rajpert-De Meyts E, Hoei-Hansen CE. Recent advances in understanding the etiology and pathogenesis of pediatric germ cell tumors. J Ped Hematol Oncol 36: 263-270, 2014.
  44. Grogg JB, Schneider K, Bode PK, Wettstein MS, Kranzbühler B, Eberli D, Sulser T, Beyer J, Hermanns T, Fankhauser CD. A systematic review of treatment outcomes in localised and metastatic spermatocytic tumors of the testis. J Cancer Res Clin Oncol. 145(12):3037-3045, 2019.
  45. Müller J, Skakkebaek NE, Parkinson MC. The spermatocytic seminoma: views on pathogenesis. Int J Androl 10: 147-156, 1987.
  46. Looijenga LH, Hersmus R, Gillis AJ, Pfundt R, Stoop HJ, van Gurp RJ, Veltman J, Beverloo HB, van Drunen E, van Kessel AG, Pera RR, Schneider DT, Summersgill B, Shipley J, McIntyre A, van der Spek P, Schoenmakers E, Oosterhuis JW. Genomic and expression profiling of human spermatocytic seminomas: primary spermatocyte as tumorigenic precursor and DMRT1 as candidate chromosome 9 gene. Cancer Res 66: 290-302, 2006.
  47. Rajpert-De Meyts E, Jacobsen GK, Bartkova J, Aubry F, Samson M, Bartek J, Skakkebaek NE. The imunohistochemical expression pattern of Chk2, p53, p19-INK4d, MAGE-A4 and other selected antigens provides new evidence for the pre-meiotic origin of spermatocytic seminoma. Histopathology 42: 217-226, 2003.
  48. Lim J, Goriely A, Turner GD, Ewen KA, Jacobsen GK, Graem N, Wilkie AO, Rajpert-De Meyts E. OCT2, SSX and SAGE1 reveal the phenotypic heterogeneity of spermatocytic seminoma reflecting distinct subpopulations of spermatogonia. J Pathol 224: 473-483, 2011.
  49. Goriely A, Hansen RM, Taylor IB, Olesen IA, Jacobsen GK, McGowan SJ, Pfeifer SP, McVean GA, Rajpert-De Meyts E, Wilkie AO. Activating mutations in FGFR3 and HRAS reveal a shared genetic origin for congenital disorders and testicular tumors. Nat Genet 41: 1247-1252, 2009.
  50. Giannoulatou E, Maher GJ, Ding Z, Gillis AJM, Dorssers LCJ, Hoischen A, Rajpert-De Meyts E, WGS500 Consortium, McVean GAT, Wilkie AOM, Looijenga LHJ, Goriely A. Whole-genome sequencing of spermatocytic tumors provides insights into the mutational processes operating in the male germline. PLoS One 2017; 12(5): e0178169. doi.org/10.1371/journal.pone.0178169
  51. Goriely A, Wilkie AO.Paternal age effect mutations and selfish spermatogonial selection: causes and consequences for human disease. Am J Hum Genet 90: 175-200, 2012
  52. Znaor A, Lortet-Tieulent J, Jemal A, Bray F. International variations and trends in testicular cancer incidence and mortality. Eur Urol. 2014 Jun;65(6):1095-106.
  53. Znaor A, Skakkebaek NE, Rajpert-De Meyts E, Kuliš T, Laversanne M, Gurney J, Sarfati D, McGlynn KA, Bray F. Global patterns in testicular cancer incidence and mortality in 2020. Int J Cancer. 2022. doi: 10.1002/ijc.33999. Epub ahead of print. PMID: 35277970.
  54. Serrano T, Chevrier C, Multigner L, Cordier S, Jégou B. International geographic correlation study of the prevalence of disorders of male reproductive health. Hum Reprod 28: 1974-1986, 2013.
  55. Gurney JK. The puzzling incidence of testicular cancer in New Zealand: what can we learn? Andrology 7(4): 394-401, 2019.
  56. Jørgensen N, Vierula M, Jacobsen R, Pukkala E, Perheentupa A, Virtanen HE, Skakkebaek NE, Toppari J. Recent adverse trends in semen quality and testis cancer incidence among Finnish men. Int J Androl 34(4 Pt 2): e37-e48, 2011.
  57. Ghazarian AA, Trabert B, Devesa SS, McGlynn KA. Recent trends in the incidence of testicular germ cell tumors in the United States. Andrology 3(1): 13-8, 2015.
  58. Møller H. Clues to the aetiology of testicular germ cell tumours from descriptive epidemiology. Eur Urol 23: 8-15, 1993.
  59. Lindhardt Johansen M, Hagen CP, Rajpert-De Meyts E, Kjærgaard S, Petersen BL, Skakkebæk NE, Main KM, Juul A. 45,X/46,XY mosaicism: phenotypic characteristics, growth, and reproductive function--a retrospective longitudinal study. J Clin Endocrinol Metab. 97: E1540-1549, 2012.
  60. Slowikowska-Hilczer J, Szarras-Czapnik M, Duranteau L, Rapp M, Walczak-Jedrzejowska R, Marchlewska K, Oszukowska E, Nordenstrom A; dsd-LIFE group. Risk of gonadal neoplasia in patients with disorders/differences of sex development. Cancer Epidemiol 69:101800. doi: 10.1016/j.canep.2020.101800, 2020.
  61. Giwercman A, Grinsted J, Hansen B, Jensen OM, Skakkebaek NE. Testicular cancer risk in boys with maldescended testis: a cohort study. J Urol 138: 1214-1216, 1987.
  62. Piltoft JS, Larsen SB, Dalton SO, Johansen C, Baker JL, Cederkvist L, Andersen I. Early life risk factors for testicular cancer: a case-cohort study based on the Copenhagen School Health Records Register. Acta Oncol. 56(2):220-224, 2017.
  63. Dieckmann KP, Rube C, Henke RP. Association of Down's syndrome and testicular cancer. J Urol 157: 1701-1704, 1997.
  64. Cook MB, Akre O, Forman D, Madigan MP, Richiardi L, McGlynn KA. A systematic review and meta-analysis of perinatal variables in relation to the risk of testicular cancer--experiences of the son. Int J Epidemiol. 39: 1605-1618, 2010.
  65. Maule M, Malavassi JL, Richiardi L. Age at puberty and risk of testicular cancer: a meta-analysis. Int J Androl. 2012 Dec;35(6):828-34. doi:10.1111/j.1365-2605.2012.01286.x. PMID: 22713104.
  66. McGlynn KA, Petrick JL, Gamborg M, Aarestrup J, Baker JL. Childhood height and risk of testicular germ cell tumors in adulthood. Int J Cancer 143(4): 767-772, 2018. 
  67. Møller H, Skakkebaek NE. Risk of testicular cancer in subfertile men: case-control study. Br Med J 318: 559-562, 1999.
  68. Jacobsen R, Bostofte E, Engholm G, Hansen J, Olsen J H, Skakkebaek NE, Møller H. Risk of testicular cancer in men with abnormal semen characteristics: cohort study. Br Med J 321: 789-792, 2000.
  69. Hanson HA, Anderson RE, Aston KI, Carrell DT, Smith KR, Hotaling JM. Subfertility increases risk of testicular cancer: evidence from population-based semen samples. Fertil Steril 105(2):322-8.e1, 2016.
  70. Fosså SD, Chen J, Schonfeld SJ, McGlynn KA, McMaster ML, Gail MH, Travis LB. Risk of contralateral testicular cancer: a population-based study of 29,515 U.S. J Natl Cancer Inst 97: 1056-1066, 2005.
  71. Hoei-Hansen CE, Holm M, Rajpert-De Meyts E, Skakkebaek NE. Histological evidence of testicular dysgenesis in contralateral biopsies of 218 patients with testicular germ cell cancer. J Pathol 200: 370-374, 2003.
  72. Zequi Sde C, da Costa WH, Santana TB, Favaretto RL, Sacomani CA, Guimaraes GC. Bilateral testicular germ cell tumours: a systematic review. BJU Int 110: 1102-1109, 2012.
  73. Watson RA, Morgan RD, Joseph J, Sivakumar S, Verrill C, Tuthill M, Sullivan M, Protheroe AS. Bilateral Testicular Germ Cell Tumors: A Case-Series From a UK-Based Tertiary Referral Center Over 19 Years. Clin Genitourinary Cancer 16: e513-e516, 2018.
  74. Dieckmann KP, Anheuser P, Sattler F, Von Kügelgen T, Matthies C, Ruf C. Sequential bilateral testicular tumours presenting with intervals of 20 years and more. BMC Urol 13: 71 (doi: 10.1186/1471-2490-13-71), 2013.
  75. Harland SJ, Cook PA, Fosså SD, Horwich A, Mead GM, Parkinson MC, Roberts JT, Stenning SP. Intratubular germ cell neoplasia of the contralateral testis in testicular cancer: defining a high risk group. J Urol 160: 1353-1357, 1998.
  76. Rud CN, Daugaard G, Rajpert-De Meyts E, Skakkebaek NE, Petersen JH, Jørgensen N. Sperm concentration, testicular volume and age predict risk of carcinoma-in-situ in contralateral testis of men with testicular germ cell cancer. J Urol 190: 2074-2080, 2013.
  77. Jørgensen N, Rajpert-De Meyts E, Main KM, Skakkebæk NE. Testicular dysgenesis syndrome comprises some but not all cases of hypospadias and impaired spermatogenesis. Int J Androl 33: 298-303, 2010.
  78. Atkin NB, Baker MC. Specific chromosome change, i(12p), in testicular tumours? Lancet. 1982 Dec 11;2(8311):1349. PubMed PMID: 6128640.
  79. Taylor-Weiner A, Zack T, O'Donnell E, Guerriero JL, Bernard B, Reddy A, Han GC, AlDubayan S, Amin-Mansour A, Schumacher SE, Litchfield K, Turnbull C, Gabriel S, Beroukhim R, Getz G, Carter SL, Hirsch MS, Letai A, Sweeney C, Van Allen EM. Genomic evolution and chemoresistance in germ-cell tumours. Nature 540(7631):114-118, 2016. PMID: 27905446.
  80. Hoff AM, Alagaratnam S, Zhao S, Bruun J, Andrews PW, Lothe RA, Skotheim RI. Identification of Novel Fusion Genes in Testicular Germ Cell Tumors. Cancer Res 76(1):108-16, 2016. doi: 10.1158/0008-5472.CAN-15-1790. PMID: 26659575.
  81. Tian Q, Frierson HFJr, Krystal GW, Moskaluk CA. Activating c-kit gene mutations in human germ cell tumors. Am J Pathol154, 1643-164, 1999.
  82. McIntyre A, Summersgill B, Grygalewicz B, Gillis AJ, Stoop J, van Gurp RJ, Dennis N, Fisher C, Huddart R, Cooper C, Clark J, Oosterhuis JW, Looijenga LH, Shipley J. Amplification and overexpression of the KIT gene is associated with progression in the seminoma subtype of testicular germ cell tumors of adolescents and adults. Cancer Res65: 8085-8089,
  83. Shen H, Shih J, Hollern DP, Wang L, Bowlby R, Tickoo SK, Thorsson V, Mungall AJ, Newton Y, Hegde AM, Armenia J, Sánchez-Vega F, Pluta J, Pyle LC, Mehra R, Reuter VE, Godoy G, Jones J, Shelley CS, Feldman DR, Vidal DO, Lessel D, Kulis T, Cárcano FM, Leraas KM, Lichtenberg TM, Brooks D, Cherniack AD, Cho J, Heiman DI, Kasaian K, Liu M, Noble MS, Xi L, Zhang H, Zhou W, ZenKlusen JC, Hutter CM, Felau I, Zhang J, Schultz N, Getz G, Meyerson M, Stuart JM; Cancer Genome Atlas Research Network, Akbani R, Wheeler DA, Laird PW, Nathanson KL, Cortessis VK, Hoadley KA. Integrated Molecular Characterization of Testicular Germ Cell Tumors. Cell Rep 23(11):3392-3406, 2018. 
  84. Hemminki K, Li X. Familial risk in testicular cancer as a clue to a heritable and environmental aetiology. Brit J Cancer 90: 1765-1770, 2004.
  85. Mucci LA, Hjelmborg JB, Harris JR, Czene K, Havelick DJ, Scheike T, Graff RE, Holst K, Möller S, Unger RH, McIntosh C, Nuttall E, Brandt I, Penney KL, Hartman M, Kraft P, Parmigiani G, Christensen K, Koskenvuo M, Holm NV, Heikkilä K, Pukkala E, Skytthe A, Adami HO, Kaprio J; Nordic Twin Study of Cancer (NorTwinCan) Collaboration. Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA 315(1):68-76, 2016. Erratum in: JAMA 315(8):822, 2016.
  86. Litchfield K, Loveday C, Levy M, Dudakia D, Rapley E, Nsengimana J, Bishop DT, Reid A, Huddart R, Broderick P, Houlston RS, Turnbull C. Large-scale Sequencing of Testicular Germ Cell Tumour (TGCT) Cases Excludes Major TGCT Predisposition Gene. Eur Urol 73(6): 828-831, 2018. 
  87. Pluta J, Pyle LC, Nead KT, Wilf R, Li M, Mitra N, Weathers B, D'Andrea K, Almstrup K, Anson-Cartwright L, Benitez J, Brown CD, Chanock S, Chen C, Cortessis VK, Ferlin A, Foresta C, Gamulin M, Gietema JA, Grasso C, Greene MH, Grotmol T, Hamilton RJ, Haugen TB, Hauser R, Hildebrandt MAT, Johnson ME, Karlsson R, Kiemeney LA, Lessel D, Lothe RA, Loud JT, Loveday C, Martin-Gimeno P, Meijer C, Nsengimana J, Quinn DI, Rafnar T, Ramdas S, Richiardi L, Skotheim RI, Stefansson K, Turnbull C, Vaughn DJ, Wiklund F, Wu X, Yang D, Zheng T, Wells AD, Grant SFA, Rajpert-De Meyts E, Schwartz SM, Bishop DT, McGlynn KA, Kanetsky PA, Nathanson KL; Testicular Cancer Consortium. Identification of 22 susceptibility loci associated with testicular germ cell tumors. Nat Commun 12(1):4487. doi: 10.1038/s41467-021-24334-y, 2021. 
  88. Rapley EA, Turnbull C, Al Olama AA, Dermitzakis ET, Linger R, Huddart RA, Renwick A, Hughes D, Hines S, Seal S, Morrison J, Nsengimana J, Deloukas P; UK Testicular Cancer Collaboration, Rahman N, Bishop DT, Easton DF, Stratton MR. A genome-wide association study of testicular germ cell tumor. Nat Genet. 41: 807-810, 2009.
  89. Kanetsky PA, Mitra N, Vardhanabhuti S, Li M, Vaughn DJ, Letrero R, Ciosek SL, Doody DR, Smith LM, Weaver J, Albano A, Chen C, Starr JR, Rader DJ, Godwin AK, Reilly MP, Hakonarson H, Schwartz SM, Nathanson KL. Common variation in KITLG and at 5q31.3 predisposes to testicular germ cell cancer. Nat Genet 41: 811-815, 2009.
  90. Turnbull C, Rapley EA, Seal S, Pernet D, Renwick A, Hughes D, Ricketts M, Linger R, Nsengimana J, Deloukas P, Huddart RA, Bishop DT, Easton DF, Stratton MR, Rahman N; UK Testicular Cancer Collaboration. Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer. Nat Genet. 42: 604-607, 2010.
  91. Dalgaard MD, Weinhold N, Edsgärd D, Silver JD, Pers TH, Nielsen JE, Jørgensen N, Juul A, Gerds TA, Giwercman A, Giwercman YL, Cohn-Cedermark G, Virtanen HE, Toppari J, Daugaard G, Jensen TS, Brunak S, Rajpert-De Meyts E, Skakkebæk NE, Leffers H, Gupta R. A genome-wide association study of men with symptoms of testicular dysgenesis syndrome and its network biology interpretation. J Med Genet. 49: 58-65, 2012.
  92. Litchfield K, Levy M, Orlando G, Loveday C, Law PJ, Migliorini G, Holroyd A, Broderick P, Karlsson R, Haugen TB, Kristiansen W, Nsengimana J, Fenwick K, Assiotis I, Kote-Jarai Z, Dunning AM, Muir K, Peto J, Eeles R, Easton DF, Dudakia D, Orr N, Pashayan N; UK Testicular Cancer Collaboration; PRACTICAL Consortium, Bishop DT, Reid A, Huddart RA, Shipley J, Grotmol T, Wiklund F, Houlston RS, Turnbull C. Identification of 19 new risk loci and potential regulatory mechanisms influencing susceptibility to testicular germ cell tumor. Nat Genet. 2017; 49(7):1133-1140.
  93. Wang Z, McGlynn KA, Rajpert-De Meyts E, Bishop DT, Chung CC, Dalgaard MD, Greene MH, Gupta R, Grotmol T, Haugen TB, Karlsson R, Litchfield K, Mitra N, Nielsen K, Pyle LC, Schwartz SM, Thorsson V, Vardhanabhuti S, Wiklund F, Turnbull C, Chanock SJ, Kanetsky PA, Nathanson KL; Testicular Cancer Consortium. Meta-analysis of five genome-wide association studies identifies multiple new loci associated with testicular germ cell tumor. Nat Genet. 2017; 49(7):1141-1147.
  94. Dolci S, Williams DE, Ernst MK, Resnick JL, Brannan CI, Lock LF, Lyman SD, Boswell HS, Donovan PJ. Requirement for mast cell growth factor for primordial germ cell survival in culture. Nature 352: 809-811, 1991.
  95. Gu Y, Runyan C, Shoemaker A, Surani A, Wylie C. Steel factor controls primordial germ cell survival and motility from the time of their specification in the allantois, and provides a continuous niche throughout their migration. Development136: 1295-1303,
  96. Matson CK, Murphy MW, Griswold MD, Yoshida S, Bardwell VJ, Zarkower D. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev Cell. 19(4):612-24, 2010.
  97. Jørgensen A, Nielsen JE, Jensen MB, Græm N, Rajpert-De Meyts E. Analysis of meiosis regulators in human gonads: a sexually dimorphic spatio-temporal expression pattern suggests involvement of DMRT1 in meiotic entry. Mol Hum Reprod 18: 523-534, 2012.
  98. Jørgensen A, Nielsen JE, Almstrup K, Toft BG, Petersen BL, Rajpert-De Meyts E. Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J Pathol. 229(4):588-598, 2013.
  99. Eini R, Dorssers LC, Looijenga LH. Role of stem cell proteins and microRNAs in embryogenesis and germ cell cancer. Int J Dev Biol 57: 319-332, 2013.
  100. Almstrup K, Nielsen JE, Mlynarska O, Jansen MT, Jørgensen A, Skakkebæk NE, Rajpert-De Meyts E. Carcinoma in situ testis displays permissive chromatin modifications similar to immature foetal germ cells. Br J Cancer 103: 1269-1276, 2010.
  101. Spiller CM, Bowles J, Koopman P. Nodal/Cripto signaling in fetal male germ cell development: implications for testicular germ cell tumors. Int J Dev Biol 57: 211-219, 2013.
  102. Jørgensen A, Macdonald J, Nielsen JE, Kilcoyne KR, Perlman S, Lundvall L, Langhoff Thuesen L, Juul Hare K, Frederiksen H, Andersson AM, Skakkebæk NE, Juul A, Sharpe RM, Rajpert-De Meyts E, Mitchell RT. Nodal Signaling Regulates Germ Cell Development and Establishment of Seminiferous Cords in the Human Fetal Testis. Cell Rep, 25(7):1924-1937, 2018.
  103. Young JC, Kerr G, Micati D, Nielsen JE, Rajpert-De Meyts E, Abud HE, Loveland KL. WNT signalling in the normal human adult testis and in male germ cell neoplasms. Hum Reprod 5(9):1991-2003, 2020, PMID: 32667987.
  104. Toppari J, Larsen JC, Christiansen P, Giwercman A, Grandjean P Guillette LJ, Jegou B, Jensen TK, Jouannet P, Keiding N, Leffers H, McLachlan JA, Meyer O, Müller J, Rajpert-De Meyts E, Scheike T, Sharpe R, Sumpter J, Skakkebaek NE. Male reproductive health and environmental xenoestrogens. Environ Health Perspect 104(Suppl 4): 741-803, 1996.
  105. van den Driesche S, Kilcoyne K, Wagner I, Rebourcet D, Boyle A, Mitchell R, McKinnell C, Macpherson S, Donat R, Shukla CJ, Joergensen A, Rajpert-De Meyts E, Skakkebæk NE, Sharpe R. Experimentally induced testicular dysgenesis syndrome originates in the masculinization programming window. JCI Insight 2017; 2(6):e91204. doi: 10.1172/jci.insight.91204.
  106. Jørgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr Oct 4:303-319, 2021.
  107. Hardell L, Bavel B, Lindström G, Eriksson M, Carlberg M. In utero exposure to persistent organic pollutants in relation to testicular cancer risk, Int J Androl 29: 228–234, 2006.
  108. McGlynn KA, Quraishi SM, Graubard BI, Weber JP, Rubertone MV, Erickson RL. Persistent organochlorine pesticides and risk of testicular germ cell tumors. J Natl Cancer Inst 100: 663–671, 2008.
  109. Bräuner EV, Lim YH, Koch T, Uldbjerg CS, Gregersen LS, Pedersen MK, Frederiksen H, Petersen JH, Coull BA, Andersson AM, Hickey M, Skakkebæk NE, Hauser R, Juul A. Endocrine Disrupting Chemicals and Risk of Testicular Cancer: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab 106(12):e4834-e4860, 2021.
  110. Rodprasert W, Toppari J, Virtanen HE. Endocrine Disrupting Chemicals and Reproductive Health in Boys and Men. Front Endocrinol (Lausanne). 2021, 12:706532. doi: 10.3389/fendo.2021.706532. PMID: 34690925.
  111. Daling JR, Doody DR, Sun X, Trabert BL, Weiss NS, Chen C, Biggs ML, Starr JR, Dey SK, Schwartz SM. Association of marijuana use and the incidence of testicular germ cell tumors. Cancer 115, 1215-1223, 2009.
  112. Gurney J, Shaw C, Stanley J, Signal V, Sarfati D. Cannabis exposure and risk of testicular cancer: a systematic review and meta-analysis. BMC Cancer 15, 897. doi: 10.1186/s12885-015-1905-6, 2015.
  113. Holm M, Hoei-Hansen CE, Rajpert-De Meyts E, Skakkebaek NE. Increased risk of carcinoma in situ in patients with testicular germ cell cancer with ultrasonic microlithiasis in the contralateral testicle. J Urol 170: 1163-1167, 2003.
  114. van Casteren NJ, Looijenga LH, Dohle GR. Testicular microlithiasis and carcinoma in situ overview and proposed clinical guideline. Int J Androl. 32: 279-287, 2009.
  115. Wang T, Liu L, Luo J, Liu T, Wei A. A Meta-Analysis of the Relationship between Testicular Microlithiasis and Incidence of Testicular Cancer. Urol J. 12(2):2057-64, 2015. PMID: 25923148.
  116. Barbonetti A, Martorella A, Minaldi E, D'Andrea S, Bardhi D, Castellini C, Francavilla F, Francavilla S. Testicular Cancer in Infertile Men With and Without Testicular Microlithiasis: A Systematic Review and Meta-Analysis of Case-Control Studies. Front Endocrinol (Lausanne10:164. 2019. doi: 10.3389/fendo.2019.00164. PMID: 30949131.
  117. Dohle GR, Elzanaty S, van Casteren NJ. Testicular biopsy: clinical practice and interpretation. Asian J Androl 14(1):88-93, 2012. doi: 10.1038/aja.2011.57. PMID: 22157985.
  118. Kliesch S, Schmidt S, Wilborn D, Aigner C, Albrecht W, Bedke J, Beintker M, Beyersdorff D, Bokemeyer C, Busch J, Classen J, de Wit M, Dieckmann KP, Diemer T, Dieing A, Gockel M, Göckel-Beining B, Hakenberg OW, Heidenreich A, Heinzelbecker J, Herkommer K, Hermanns T, Kaufmann S, Kornmann M, Kotzerke J, Krege S, Kristiansen G, Lorch A, Müller AC, Oechsle K, Ohloff T, Oing C, Otto U, Pfister D, Pichler R, Recken H, Rick O, Rudolph Y, Ruf C, Schirren J, Schmelz H, Schmidberger H, Schrader M, Schweyer S, Seeling S, Souchon R, Winter C, Wittekind C, Zengerling F, Zermann DH, Zillmann R, Albers P. Management of Germ Cell Tumours of the Testis in Adult Patients. German Clinical Practice Guideline Part I: Epidemiology, Classification, Diagnosis, Prognosis, Fertility Preservation, and Treatment Recommendations for Localized Stages. Urol Int. 2021;105(3-4):169-180. doi: 10.1159/000510407.
  119. McLachlan RI, Rajpert-De Meyts E, Hoei-Hansen CE, De Kretser DM, Skakkebaek NE. Histological evaluation of the human testis: approaches to optimizing the clinical value of the assessment: mini review, Hum Reprod 22: 2–16, 2007.
  120. van Casteren NJ, de Jong J, Stoop H, Steyerberg EW, de Bekker-Grob EW, Dohle GR, Oosterhuis JW & Looijenga LH.Evaluation of testicular biopsies for carcinoma in situ: immunohistochemistry is mandatory. Int J Androl 32, 666-674, 2009.
  121. Kier MGG, Lauritsen J, Almstrup K,Mortensen MS, Toft BG, Rajpert-De Meyts E,  Skakkebæk NE, Rørth M, von der Maase H, Agerbæk M, Holm NV, Andersen KK,  Dalton SO, Johansen C, Daugaard G. Screening for carcinoma in situ in the contralateral testicle in patients with testicular cancer: a population-based study. Ann Oncol 15; 26: 737-742, 2015.
  122. Rajpert-De Meyts E, Jørgensen N, Petersen JH, Almstrup K, Aksglaede L, Lauritsen J, Rørth M, Daugaard G, Skakkebaek NE. Optimized detection of germ cell neoplasia in situ in contralateral biopsy reduces the risk of second testis cancer. BJU Int. 2022 doi: 10.1111/bju.15774. Epub ahead of print. PMID: 35575005.
  123. Albers P, Albrecht W, Algaba F, Bokemeyer C, Cohn-Cedermark G, Fizazi K, Horwich A, Laguna MP, Nicolai N, Oldenburg J; European Association of Urology. Guidelines on testicular cancer:2015 update. Eur Urol. 68: 1054-1068, 2015.  
  124. Gilligan T, Lin DW, Aggarwal R, Chism D, Cost N, Derweesh IH, Emamekhoo H, Feldman DR, Geynisman DM, Hancock SL, LaGrange C, Levine EG, Longo T, Lowrance W, McGregor B, Monk P, Picus J, Pierorazio P, Rais-Bahrami S, Saylor P, Sircar K, Smith DC, Tzou K, Vaena D, Vaughn D, Yamoah K, Yamzon J, Johnson-Chilla A, Keller J, Pluchino LA. Testicular Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 17(12):1529-1554, 2019, doi: 10.6004/jnccn.2019.0058. PMID: 31805523.
  125. Bagrodia A, Pierorazio P, Singla N, Albers P. The Complex and Nuanced Care for Early-stage Testicular Cancer: Lessons from the European Association of Urology and American Urological Association Testis Cancer Guidelines. Eur Urol 77(2):139-141, 2020. doi: 10.1016/j.eururo.2019.10.003, PMID: 31672465.
  126. Dieckmann KP, Kulejewski M, Pichlmeier U, Loy V. Diagnosis of contralateral testicular intraepithelial neoplasia (TIN) in patients with testicular germ cell cancer: systematic two-site biopsies are more sensitive than a single random biopsy. Eur Urol 51:175–183, 2007.
  127. Dieckmann KP, Heinemann V, Frey U, Pichlmeier U. German Testicular Cancer Study Group. How harmful is contralateral testicular biopsy? An analysis of serial imaging studies and a prospective evaluation of surgical complications. Eur Urol. 48: 662-672, 2005.
  128. Dieckmann KP, Loy V. False-negative biopsies for the diagnosis of testicular intraepithelial neoplasia (TIN)--an update. Eur Urol. 43: 516-521, 2003.
  129. Rajpert-De Meyts E, Grigor KM, Skakkebæk NE. Histopathological evaluation of testicular biopsy. In: Endocrinology of the Testis and Male Reproduction. M. Simoni, I. Huhtaniemi (Editors). Springer International Publishing, 2017, pp. 1-20, doi:10.1007/978-3-319-29456-8_20-1.
  130. Petersen PM, Skakkebaek NE, Vistisen K, Rørth M, Giwercman A. Semen quality and reproductive hormones before orchiectomy in men with testicular cancer. J Clin Oncol 17: 941-947,
  131. Del Giudice F, Kasman AM, De Berardinis E, Busetto GM, Belladelli F, Eisenberg ML. Association between male infertility and male-specific malignancies: systematic review and meta-analysis of population-based retrospective cohort studies. Fertil Steril 114(5):984-996, 2020. doi: 10.1016/j.fertnstert.2020.04.042. PMID: 32709378.
  132. Giwercman A, Marks A, Skakkebaek NE. Carcinoma-in-situ germ-cells exfoliated from seminiferous epithelium into seminal fluid. Lancet. 1(8584): 530, 1988.
  133. Hoei-Hansen CE, Carlsen E, Jørgensen N, Leffers H Skakkebæk NE, Rajpert-De Meyts E. Towards a non-invasive method for early detection of testicular neoplasia in semen samples by identification of fetal germ cell-specific markers. Hum Reprod 22: 167-173, 2007.
  134. van Casteren NJ, Stoop H, Dohle GR, de Wit R, Oosterhuis JW, Looijenga LHJ. Noninvasive detection of testicular carcinoma in situ in semen using OCT3/4, Eur Urol 54: 153–158, 2008.
  135. Almstrup K, Lippert M, Mogensen HO, Nielsen JE, Hansen JD, Daugaard G, Jørgensen N, Foged NT, Skakkebæk NE, Rajpert-De Meyts E. Screening of subfertile men for testicular carcinoma in situ by an automated image analysis-based cytological test of the ejaculate. Int J Androl 34: e21-e31, 2011.
  136. Radtke A, Cremers JF, Kliesch S, Riek S, Junker K, Mohamed SA, Anheuser P, Belge G, Dieckmann KP. Can germ cell neoplasia in situ be diagnosed by measuring serum levels of microRNA371a-3p? J Cancer Res Clin Oncol. 143(11): 2383-2392, 2017.
  137. Pelloni M, Coltrinari G, Paoli D, Pallotti F, Lombardo F, Lenzi A, Gandini L. Differential expression of miRNAs in the seminal plasma and serum of testicular cancer patients. Endocrine. 2017 Sep;57(3):518-527. doi: 10.1007/s12020-016-1150-z. Epub 2016 Oct 28. PMID: 27796811.
  138. Boellaard WPA, Gillis AJM, van Leenders GJLH, Stoop H, van Agthoven T, Dorssers LCJ, Dinkelman-Smit M, Boormans JL, Looijenga LHJ. Cellular origin of microRNA-371a-3p in healthy males based on systematic urogenital tract tissue evaluation. Andrology 7(4):463-468, 2019. doi: 10.1111/andr.12595. PMID: 30786164.
  139. Radtke A, Dieckmann KP, Grobelny F, Salzbrunn A, Oing C, Schulze W, Belge G. Expression of miRNA-371a-3p in seminal plasma and ejaculate is associated with sperm concentration. Andrology 7(4):469-474, 2019. doi: 10.1111/andr.12664. PMID: 31310058.
  140. Gilligan TD, Seidenfeld J, Basch EM, Einhorn LH, Fancher T, Smith DC, Stephenson AJ, Vaughn DJ, Cosby R, Hayes DF; American Society of Clinical Oncology. American Society of Clinical Oncology Clinical Practice Guideline on uses of serum tumor markers in adult males with germ cell tumors. J Clin Oncol 28:3388-3404, 2010.
  141. Gitlin D, Perricelli A: Synthesis of serum albumin, prealbumin, alpha-fetoprotein, 1-antitrypsin and transferrin by the human yolk sac, Nature 228:995–996, 1970.
  142. von Eyben FE, de Graaff WE, Marrink J, Blaabjerg O, Sleijfer DT, Koops HS, Oosterhuis JW, Petersen PH, van Echten-Arends J, de Jong B. Serum lactate dehydrogenase isoenzyme-1 activity in patients with testicular germ cell tumours correlates with the total number of copies of the short arm of chromosome 12 in the tumour. Mol Gen Genet 235: 140-147, 1991.
  143. Bandak M, Jørgensen N, Juul A, Lauritsen J, Gundgaard Kier MG, Mortensen MS, et al. Preorchiectomy Leydig cell dysfunction in patients with testicular cancer. Clin Genitourin Cancer 15(1):e37–43, 2016. doi:10.1016/j.clgc.2016. 07.006
  144. Palmer RD, Murray MJ, Saini HK, et al. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Research 70, 2911-2923, 2010.
  145. Gillis AJ, Rijlaarsdam MA, Eini R, et al. Targeted serum miRNA (TSmiR) test for diagnosis and follow-up of (testicular) germ cell cancer patients: a proof of principle. Mol Oncology 7, 1083-1092, 2013.
  146. Dieckmann KP, Radtke A, Geczi L, Matthies C, Anheuser P, Eckardt U, Sommer J, Zengerling F, Trenti E, Pichler R, Belz H, Zastrow S, Winter A, Melchior S, Hammel J, Kranz J, Bolten M, Krege S, Haben B, Loidl W, Ruf CG, Heinzelbecker J, Heidenreich A, Cremers JF, Oing C, Hermanns T, Fankhauser CD, Gillessen S, Reichegger H, Cathomas R, Pichler M, Hentrich M, Eredics K, Lorch A, Wülfing C, Peine S, Wosniok W, Bokemeyer C, Belge G. Serum Levels of MicroRNA-371a-3p (M371 Test) as a New Biomarker of Testicular Germ Cell Tumors: Results of a Prospective Multicentric Study. J Clin Oncol 37(16):1412-1423, 2019. doi: 10.1200/JCO.18.01480.
  147. Piao J, Lafin JT, Scarpini CG, Nuño MM, Syring I, Dieckmann KP, Belge G, Ellinger J, Amatruda JF, Bagrodia A, Coleman N, Krailo MD, Frazier AL, Murray MJ. A Multi-institutional Pooled Analysis Demonstrates That Circulating miR-371a-3p Alone is Sufficient for Testicular Malignant Germ Cell Tumor Diagnosis. Clin Genitourin Cancer. 2021; 19(6):469-479. doi: 10.1016/j.clgc.2021.08.006..
  148. Aganovic L, Cassidy F. Imaging of the scrotum. Radiol Clin North Am. 50: 1145-1165, 2012.
  149. Cantisani V, Di Leo N, Bertolotto M, Fresilli D, Granata A, Polti G, Polito E, Pacini P, Guiban O, Del Gaudio G, Dolcetti V, DʼAndrea V, Di Pierro GB, Verrengia M, Drudi FM, Catalano C. Role of multiparametric ultrasound in testicular focal lesions and diffuse pathology evaluation, with particular regard to elastography: Review of literature. Andrology 9(5):1356-1368, 2021. doi: 10.1111/andr.13067.
  150. Tsili AC, Argyropoulou MI, Astrakas LG, Ntoulia EA, Giannakis D, Sofikitis N, Tsampoulas K. Dynamic contrast-enhanced subtraction MRI for characterizing intratesticular mass lesions. AJR Am J Roentgenol. 200: 578-585. 2013.
  151. Honecker F, Aparicio J, Berney D, Beyer J, Bokemeyer C, Cathomas R, Clarke N, Cohn-Cedermark G, Daugaard G, Dieckmann KP, Fizazi K, Fosså S, Germa-Lluch JR, Giannatempo P, Gietema JA, Gillessen S, Haugnes HS, Heidenreich A, Hemminki K, Huddart R, Jewett MAS, Joly F, Lauritsen J, Lorch A, Necchi A, Nicolai N, Oing C, Oldenburg J, Ondruš D, Papachristofilou A, Powles T, Sohaib A, Ståhl O, Tandstad T, Toner G, Horwich A. ESMO Consensus Conference on testicular germ cell cancer: diagnosis, treatment and follow-up. Ann Oncol 29(8):1658-1686, 2018. doi: 10.1093/annonc/mdy217.
  152. Daugaard G, Gundgaard MG, Mortensen MS, Agerbaek M, Holm NV, Rorth M, et a Surveillance for stage I nonseminoma testicular cancer: outcomes and long-term follow-up in a population-based cohort. J Clin Oncol 32(34): 3817-23, 2014.
  153. Verrill C, Yilmaz A, Srigley JR, et al. et Members of the International Society of Urological Pathology Testicular Tumor Panel. Reporting and Staging of Testicular Germ Cell Tumors: The International Society of Urological Pathology (ISUP) Testicular Cancer Consultation Conference Recommendations. Am J Surg Pathol 41, e22-e32, 2017.
  154. Sobin LH, Gospodarowicz M, Wittekind C (eds). TNM Classification of Malignant Tumours, UICC International Union Against Cancer 7th ed. Wiley-Blackwell, pp 249-254, 2009.
  155. Chresta CM, Masters JWR, Hickman JA. Hypersensitivity of human testicular tumors to etoposide-induced apoptosis is associated with functional p53 and a high bax:bcl-2 ratio. Cancer Res 56: 1834-1841, 1996.
  156. Timmerman DM, Eleveld TF, Gillis AJM, Friedrichs CC, Hillenius S, Remmers TL, Sriram S, Looijenga LHJ. The Role of TP53in Cisplatin Resistance in Mediastinal and Testicular Germ Cell Tumors. Int J Mol Sci. 22(21):11774, 2021. doi: 10.3390/ijms222111774.
  157. King J, Adra N, Einhorn LH. Testicular Cancer: Biology to Bedside. Cancer Res. 2021; 81(21):5369-5376. doi: 10.1158/0008-5472.CAN-21-1452.
  158. Mortensen MS, Gundgaard MG, Daugaard G. Treatment options for carcinoma in situ testis. Int J Androl. 34(4 Pt 2): e32-e36, 2011.
  159. Giwercman A, von der Maase H, Berthelsen JG, Rorth M, Bertelsen A, Skakkebaek NE. Localized irradiation of testes with carcinoma in situ: effects on Leydig cell function and eradication of malignant germ cells in 20 patients. J Clin Endocrinol Metab 73: 596-603, 1991.
  160. Bang AK, Petersen JH, Petersen PM, Andersson AM, Daugaard G, Jørgensen N. Testosterone production is better preserved after 16 than 20 Gray irradiation treatment against testicular carcinoma in situ cells. Int J Radiat Oncol Biol Phys.75: 672-676, 2009.
  161. Dieckmann KP, Wilken S, Loy V, Matthies C, Kleinschmidt K, Bedke J, Martinschek A, Souchon R, Pichlmeier U, Kliesch S. Treatment of testicular intraepithelial neoplasia (intratubular germ cell neoplasia unspecified) with local radiotherapy or with platinum-based chemotherapy: a survey of the German Testicular Cancer Study Group. Ann Oncol 24(5):1332-7, 2013. doi: 10.1093/annonc/mds628.
  162. Fosså SD, Aass N, Heilo A, Daugaard G, E Skakkebaek N, Stenwig AE, Nesland JM, Looijenga LH, Oosterhuis JW. Testicular carcinoma in situ in patients with extragonadal germ-cell tumours: the clinical role of pretreatment biopsy. Ann Oncol 14: 1412-1418, 2003.
  163. Blok JM, Groot HJ, Huele EH, de Wit R, Horenblas S, Nuver J, Groenewegen G, Bosch JLHR, Witjes JA, Tromp JM, de Brouwer PJM, van den Berg HA, Vanneste BGL, Smilde TJ, Aarts MJB, Gietema JA, Meijer RP, Schaapveld M.  Dose-Dependent Effect of Platinum-Based Chemotherapy on the Risk of Metachronous Contralateral Testicular Cancer. J Clin Oncol 39: 319-327, 2021.
  164. Hellesnes R, Myklebust TÅ, Bremnes RM, Karlsdottir Á, Kvammen Ø, Negaard HFS, Tandstad T, Wilsgaard T, Fosså SD, Haugnes HS. Metachronous Contralateral Testicular Cancer in the Cisplatin Era: A Population-Based Cohort Study. J Clin Oncol 39(4):308-318, 2021. PMID: 33356420.
  165. Schrader M, Müller M, Sofikitis N, Straub B, Krause H, Miller K. "Onco-TESE": testicular sperm extraction in azoospermic cancer patients before chemotherapy-new guidelines? Urology 61: 421-425, 2003.
  166. Suzuki K, Shin T, Shimomura Y, Iwahata T, Okada H. Spermatogenesis in tumor-bearing testes in germ cell testicular cancer patients. Hum Reprod 30(12):2853-2858, 2015.
  167. Beyer J, Albers P, Altena R, Aparicio J, Bokemeyer C, Busch J, et al. Maintaining success, reducing treatment burden, focusing on survivorship: highlights from the third European consensus conference on diagnosis and treatment of germ-cell cancer. Ann Oncol 24(4): 878-88, 2013.
  168. Nichols CR, Roth B, Albers P, Einhorn LH et al. Active surveillance is the preferred approach to clinical stage I testicular cancer. J Clin Oncol 31: 3490-3493, 2013.
  169. Heidenreich A, Paffenholz P, Nestler T, Pfister D. Primary and Postchemotherapy Retroperitoneal Lymphadenectomy for Testicular Cancer. Oncol Res Treat 41(6):370-378, 2018. doi: 10.1159/000489508.
  170. Mortensen MS, Lauritsen J, Gundgaard MG, Agerbaek M, Holm NV, Christensen IJ, et al. A nationwide cohort study of stage I seminoma patients followed on a surveillance program. Eur Urol66(6):1172–8, 2014. doi:10.1016/j.eururo.2014.07.001.
  171. Tandstad T, Ståhl O, Håkansson U, et al. One course of adjuvant BEP in clinical stage I nonseminoma mature and expanded results from the SWENOTECA group. Ann Oncol 25, 2167-2172, 2014.
  172. Oldenburg J, Berney DM, Bokemeyer C, Climent MA, Daugaard G, Gietema JA, De Giorgi U, Haugnes HS, Huddart RA, Leão R, Sohaib A, Gillessen S, Powles T; ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org; EURACAN. Testicular seminoma and nonseminoma: ESMO-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 33(4):362-375, 2022. doi: 10.1016/j.annonc.2022.01.002. PMID: 35065204.
  173. Beyer J, Collette L, Sauvé N, Daugaard G, Feldman DR, Tandstad T, Tryakin A, Stahl O, Gonzalez-Billalabeitia E, De Giorgi U, Culine S, de Wit R, Hansen AR, Bebek M, Terbuch A, Albany C, Hentrich M, Gietema JA, Negaard H, Huddart RA, Lorch A, Cafferty FH, Heng DYC, Sweeney CJ, Winquist E, Chovanec M, Fankhauser C, Stark D, Grimison P, Necchi A, Tran B, Heidenreich A, Shamash J, Sternberg CN, Vaughn DJ, Duran I, Bokemeyer C, Patrikidou A, Cathomas R, Assele S, Gillessen S; International Germ Cell Cancer Classification Update Consortium. Survival and New Prognosticators in Metastatic Seminoma: Results From the IGCCCG-Update Consortium. J Clin Oncol 39(14):1553-1562, 2021. doi: 10.1200/JCO.20.03292. PMID: 33729863.
  174. Gillessen S, Sauvé N, Collette L, Daugaard G, de Wit R, Albany C, Tryakin A, Fizazi K, Stahl O, Gietema JA, De Giorgi U, Cafferty FH, Hansen AR, Tandstad T, Huddart RA, Necchi A, Sweeney CJ, Garcia-Del-Muro X, Heng DYC, Lorch A, Chovanec M, Winquist E, Grimison P, Feldman DR, Terbuch A, Hentrich M, Bokemeyer C, Negaard H, Fankhauser C, Shamash J, Vaughn DJ, Sternberg CN, Heidenreich A, Beyer J; International Germ Cell Cancer Classification Update Consortium. Predicting Outcomes in Men With Metastatic Nonseminomatous Germ Cell Tumors (NSGCT): Results From the IGCCCG Update Consortium. J Clin Oncol 39(14):1563-1574, 2021. doi: 10.1200/JCO.20.03296. PMID: 33822655.
  175. Borer JG, Tan PE, Diamond DA. The spectrum of Sertoli cell tumors in children. Urol Clin North Am 27: 529-541, 2000.
  176. Harms D, Kock LR. Testicular juvenile granulosa cell and Sertoli cell tumours: a clinicopathological study of 29 cases from the Kiel Paediatric Tumour Registry. Virchows Arch 430: 301-309, 1997.
  177. Kaplan GW, Cromie WJ, Kelalis PP, Silber I, Tank ESJr. Gonadal stromal tumors: a report of prepubertal tumor registry. J Urol 136: 300-302, 1986.
  178. Idrees MT, Ulbright TM, Oliva E, et al. et Members of the International Society of Urological Pathology Testicular Tumour Panel. The World Health Organization 2016 classification of testicular non-germ cell tumours: a review and update from the International Society of Urological Pathology Testis Consultation Panel. Histopathology 70: 513-521, 2017.
  179. Iczkowski KA, Bostwick DG, Roche PC, Cheville JC. Inhibin A is a sensitive and specific marker for testicular sex cord-stromal tumors. Mod Pathol 11: 774-779, 1998.
  180. Toppari, J, Kaipia A, Kaleva M et al. Inhibin gene expression in a large cell calcifying Sertoli cell tumour and serum inhibin and activin levels. APMIS 106: 101-113, 1998.
  181. Henley JD, Young RH, Ulbright TM. Malignant Sertoli cell tumors of the testis: a study of 13 examples of a neoplasm frequently misinterpreted as seminoma. Am J Surg Pathol 26: 541-550, 2002.
  182. Kommoss F, Oliva E, Bittinger F, Kirkpatrick CJ, Amin MB, Bhan AK, Young RH, Scully RE. Inhibin-alpha CD99, HEA125, PLAP, and chromogranin immunoreactivity in testicular neoplasms and the androgen insensitivity syndrome. Hum Pathol 31: 1055-1061, 2000.
  183. Rey R, Sabourin JC, Venara M, Long WQ, Jaubert F, Zeller WP, Duvillard P, Chemes H, Bidart JM. Anti-Mullerian hormone is a specific marker of Sertoli- and granulosa-cell origin in gonadal tumors. Hum Pathol 31: 1202-1208, 2000.
  184. Ketola I, Pentikainen V, Vaskivuo T, Ilvesmaki V, Herva R, Dunkel L, Tapanainen JS, Toppari J, Heikinheimo M. Expression of transcription factor GATA-4 during human testicular development and disease. J Clin Endocrinol Metab 85: 3925-3931, 2000.
  185. Lottrup G, Nielsen JE, Skakkebæk NE, Juul A, Rajpert-De Meyts E. Abundance of DLK1, differential expression of CYP11B1, CYP21A2, MC2R and lack of INSL3 distinguish testicular adrenal rest tumours from Leydig cell tumours. Eur J Endocrinol 172: 491-499, 2015.
  186. Holm M, Rajpert-De Meyts E, Andersson A-M, and Skakkebæk NE. Leydig cell micronodules are a common finding in testicular biopsies from men with impaired spermatogenesis and are associated with decreased Testosterone/LH ratio. J Pathol 199: 378-386, 2003.
  187. Lottrup G, Nielsen JE, Maroun LL, Møller LMA, Yassin M, Leffers H, Skakkebæk NE, Rajpert-De Meyts E. Expression patterns of DLK1 and INSL3 identify stages of Leydig cell differentiation during normal development and in testicular pathologies, including testicular cancer and Klinefelter syndrome. Hum Reprod 29: 1637-1650, 2014.
  188. Fankhauser CD, Grogg JB, Hayoz S, et al. Risk Factors and Treatment Outcomes of 1,375 Patients with Testicular Leydig Cell Tumors: Analysis of Published Case Series Data. J Urol 203:949-956, 2020.
  189. Sørensen RR, Johannsen TH, Skakkebaek NE, Rajpert-De Meyts E. Leydig cell clustering and Reinke crystal distribution in relation to hormonal function in adult patients with testicular dysgenesis syndrome (TDS) including cryptorchidism. Hormones 15: 518-526, 2016.
  190. Lottrup G, Belling K, Leffers H, Nielsen JE, Dalgaard MD, Juul A, Skakkebæk NE, Brunak S, Rajpert-De Meyts E. Comparison of global gene expression profiles of microdissected human foetal Leydig cells with their normal and hyperplastic adult equivalents. Mol Hum Reprod 23 (5): 339-354, 2017.
  191. Rich MA, Keating MA. Leydig cell tumors and tumors associated with congenital adrenal hyperplasia. Urol Clin North Am 27: 519-528, 2000.
  192. Pozza C, Pofi R, Tenuta M, Tarsitano MG, Sbardella E, Fattorini G, Cantisani V, Lenzi A, Isidori AM, Gianfrilli D; TESTIS UNIT. Clinical presentation, management and follow-up of 83 patients with Leydig cell tumors of the testis: a prospective case-cohort study. Hum Reprod 34(8):1389-1403, 2019. doi: 10.1093/humrep/dez083. PMID: 31532522.
  193. Shenker A, Laue L, Kosugi S, Merendino JJ Jr, Minegishi T, Cutler GB Jr. A constitutively activating mutation of the luteinizing hormone receptor in familial male precocious puberty. Nature 365: 652-654, 1993.
  194. Liu G, Duranteau L, Carel JC, Monroe J, Doyle DA, Shenker A. Leydig-cell tumors caused by an activating mutation of the gene encoding the luteinizing hormone receptor. N Engl J Med 341: 173-176, 1999.
  195. Mortensen LJ, Blomberg Jensen M, Christiansen P, Rønholt AM, Jørgensen A, Frederiksen H, Nielsen JE, Loya AC, Grønkær Toft B, Skakkebæk NE, Rajpert-De Meyts E, Juul A. Germ cell neoplasia in situ and preserved fertility despite suppressed gonadotropins in a patient with testotoxicosis. J Clin Endocrinol Metab 102: 4411–4416, 2017.
  196. Iiri T, Herzmark P, Nakamoto JM, van Dop C, Bourne HR. Rapid GDP release from Gs alpha in patients with gain and loss of endocrine function. Nature 371: 164-168, 1994.
  197. Fragoso MC, Latronico AC, Carvalho FM, Zerbini MC, Marcondes JA, Araujo LM, Lando VS, Frazzatto ET, Mendonca BB, Villares SM. Activating mutation of the stimulatory G protein (gsp) as a putative cause of ovarian and testicular human stromal Leydig cell tumors. J Clin Endocrinol Metab 83: 2074-2081, 1998.
  198. Themmen APN, Brunner HG. Luteinizing hormone receptor mutations and sex differentiation. Eur J Endocrinol 134: 533-540, 1996.
  199. Carvajal-Carmona LG, Alam NA, Pollard PJ, et al: Adult Leydig cell tumors of the testis caused by germline fumarate hydratase mutations, J Clin Endocrinol Metab 91:3071–3075, 2006.
  200. Mieritz MG, Raket LL, Hagen CP, Nielsen JE, Talman ML, Petersen JH, Sommer SH, Main KM, Jørgensen N, Juul A. A Longitudinal Study of Growth, Sex Steroids, and IGF-1 in Boys With Physiological Gynecomastia. J Clin Endocrinol Metab 100:3752-3759, 2015.
  201. Kanakis GA, Nordkap L, Bang AK, Calogero AE, Bártfai G, Corona G, Forti G, Toppari J, Goulis DG, Jørgensen N. EAA clinical practice guidelines - gynecomastia evaluation and management. Andrology 7:778-793, 2019.
  202. Kim I, Young RH, Scully RE. Leydig cell tumors of the testis. A clinicopathologic study of 40 cases and review of the literature. Am J Surg Pathol 9: 177-192, 1985.
  203. Rushton HG, Belman AB. Testis-sparing surgery for benign lesions of the prepubertal testis. Urol Clin North Am 20: 27-37, 1993.
  204. Bozzini G, Picozzi S, Gadda F, Colombo R, Decobelli O, Palou J, Colpi G, Carmignani L. Long-term follow-up using testicle-sparing surgery for Leydig cell tumor. Clin Genitourin Cancer11(3): 321-324, 2013.
  205. Fankhauser CD, Roth L, Kranzbühler B, Eberli D, Bode P, Moch H, Oliveira P, Ramani V, Beyer J, Hermanns T. The Role of Frozen Section Examination During Inguinal Exploration in Men with Inconclusive Testicular Tumors: A Systematic Review and Meta-analysis. Eur Urol Focus 7(6):1400-1402, 2021. doi: 10.1016/j.euf.2020.06.019. PMID: 32684510.
  206. Cheville JC, Sebo TJ, Lager DJ, Bostwick DG, Farrow GM. Leydig cell tumor of the testis: a clinicopathologic, DNA content, and MIB-1 comparison of nonmetastasizing and metastasizing tumors. Am J Surg Pathol 22: 1361-1367, 1998.
  207. Calaway AC, Tachibana I, Masterson TA Foster RS, Einhorn LH, Cary C. Oncologic outcomes following surgical management of clinical stage II sex cord stromal tumors. Urology 127: 74-79, 2019, PMID: 30807775.
  208. Hamwi GJ, Gwinup G, Mostow JH, Besch PK. Activation of testicular adrenal rest tissue by prolonged excessive ACTH production. J Clin Endocrinol Metab 23: 861-869, 1963.
  209. Engels M, Span PN, van Herwaarden AE, Sweep FCGJ, Stikkelbroeck NMML, Claahsen-van der Grinten HL.Testicular Adrenal Rest Tumors: Current Insights on Prevalence, Characteristics, Origin, and Treatment. Endocr Rev 40:973-987, 2019.
  210. Venara M, Rey R, Bergada I, Mendilaharzu H, Campo S, Chemes H. Sertoli cell proliferations of the infantile testis: an intratubular form of Sertoli cell tumor? Am J Surg Pathol 25: 1237-1244, 2001.
  211. Young RH, Koelliker DD, Scully RE. Sertoli cell tumors of the testis, not otherwise specified. Am J Surg Pathol 22:709-721, 1998.
  212. Perrone F, Bertolotti A, Montemurro G, Paolini B, Pierotti MA, Colecchia M. Frequent mutation and nuclear localization of β-catenin in Sertoli cell tumors of the testis. Am J Surg Pathol 38(1):66-71, 2014.
  213. Giglio M, Medica M, De Rose AF, Germinale F, Ravetti JL, Carmignani G. Giglio M, Medica M, De Rose AF, Germinale F, Ravetti JL, Carmignani G. Testicular Sertoli cell tumours and relative sub-types. Analysis of clinical and prognostic features. Urol Int 70: 205-210, 2003.
  214. Gourgari E, Saloustros E, Stratakis CA. Large-cell calcifying Sertoli cell tumors of the testes in pediatrics. Curr Opin Pediatr 24:518-22, 2012.
  215. Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL. The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine 64: 270-283, 1985.
  216. Proppe KH, Scully RE. Large-cell calcifying Sertoli cell tumor of the testis. Am J Clin Pathol 74: 607-619, 1980.
  217. Washecka R, Dresner MI, Honda SA. Testicular tumors in Carney’s complex. J Urol 167: 1299-1302, 2002.
  218. Kratzer SS, Ulbright TM, Talerman A, Srigley JR, Roth LM, Wahle GR, Moussa M, Stephens JK, Millos A, Young RH. Large cell calcifying Sertoli cell tumor of the testis. Contrasting features of six malignant and six benign tumors and a review of literature. Am J Surg Pathol 21: 1271-1280, 1997.
  219. Stratakis CA, Carney JA, Lin JP, Papanicolau DA, Karl M, Kastner DL, Pras E, Chrousos GP. Carney complex, a familial multiple neoplasia and lentiginosis syndrome. Analysis of 11 kindreds and linkage to the short arm of chromosome 2. J Clin Invest 97: 699-705, 1996.
  220. Casey M, Mah C, Merliss AD, Kirschner LS, Taymans SE, Denio AE, Korf B, Irvine AD, Hughes A, Carney JA, Stratakis CA, Basson CT. Identification of a novel genetic locus for familial cardiac myxomas and Carney complex. Circulation 98: 2560-2566, 1998.
  221. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients the Carney complex. Nat Genet 26: 89-92, 2000.
  222. Casey M, Vaughan CJ, He J, Hatcher CJ, Winter JM, Weremowicz S, Montgomery K, Kucherlapati R, Morton CC, Basson CT. Mutations in the protein kinase A R1alpha regulatory subunit cause familial cardiac myxomas and Carney complex. J Clin Invest 106: R31-8, 2000.
  223. Libé R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin-Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA. Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab. 96(1):E208-214, 2011.
  224. Ham S, Meachem SJ, Choong CS, Charles AK, Baynam GS, Jones TW, Samarajeewa NU, Simpson ER, Brown KA. Overexpression of aromatase associated with loss of heterozygosity of the STK11 gene accounts for prepubertal gynecomastia in boys with Peutz-Jeghers syndrome. J Clin Endocrinol Metab 98, E1979-E1987, 2013.
  225. Hemminki A, Markie D, Tomlison I, Avizienyte E, Roth S, Loukola A, Bignell G, Warren W, Aminoff M, Hoglund P, Jarvinen H, Kristo P, Pelin K, Ridanpaa M, Salovaara R, Toro T, Bodmer W, Olschwang S, Olsen AS, Stratton MR, de la Chapelle A, Aaltonen LA. A serine/threonine kinase gene defective in Peutz-Jegher syndrome. Nature 391: 184-187, 1998.
  226. Brown B, Ram A, Clayton P, et al: Conservative management of bilateral Sertoli cell tumors of the testicle in association with the Carney complex: a case report, J Pediatr Surg 42:E13–15, 2007.
  227. Grogg J, Schneider K, Bode PK, Kranzbühler B, Eberli D, Sulser T, Lorch A, Beyer J, Hermanns T, Fankhauser CD. Sertoli Cell Tumors of the Testes: Systematic Literature Review and Meta-Analysis of Outcomes in 435 Patients. Oncologist 25(7):585-590, 2020. PMID: 32043680.
  228. Hofmann M, Schlegel PG, Hippert F, Schmidt P, von-Schweinitz D, Leuschner I, Göbel U, Calaminus G, Schneider DT; MAKEI study group. Testicular sex cord stromal tumors: Analysis of patients from the MAKEI study. Pediatr Blood Cancer. 60(10):1651-1655, 2013.
  229. Kao CS, Cornejo KM, Ulbright TM, Young RH. Juvenile granulosa cell tumors of the testis: a clinicopathologic study of 70 cases with emphasis on its wide morphologic spectrum. Am J Surg Pathol 39(9):1159-69, 2015.
  230. Grogg JB, Schneider K, Bode PK, Kranzbühler B, Eberli D, Sulser T, Beyer J, Lorch A, Hermanns T, Fankhauser CD. Risk factors and treatment outcomes of 239 patients with testicular granulosa cell tumors: a systematic review of published case series data. J Cancer Res Clin Oncol. 146(11):2829-2841, 2020. PMID: 32719989.
  231. Boyer A, Paquet M, Laguë MN, Hermo L, Boerboom D. Dysregulation of WNT/CTNNB1 and PI3K/AKT signaling in testicular stromal cells causes granulosa cell tumor of the testis. Carcinogenesis. 30(5):869-878, 2009.
  232. Kalfa N, Ecochard A, Patte C, Duvillard P, Audran F, Pienkowski C, Thibaud E, Brauner R, Lecointre C, Plantaz D, Guedj AM, Paris F, Baldet P, Lumbroso S, Sultan C. Activating mutations of the stimulatory G protein in juvenile ovarian granulosa cell tumors: a new prognostic factor? J Clin Endocrinol Metab. 91(5):1842-1847, 2006.
  233. Jimenez-Quintero LP, Ro JY, Zavala-Pompa A, Amin MB, Tetu B, Ordonez NG, Ayala AG. Granulosa cell tumor of the adult testis: a clinicopathologic study of seven cases and a review of the literature. Hum Pathol 24: 1120-1125, 1993.
  234. Shah SP, Köbel M, Senz J, Morin RD, Clarke BA, Wiegand KC, Leung G, Zayed A, Mehl E, Kalloger SE, Sun M, Giuliany R, Yorida E, Jones S, Varhol R, Swenerton KD, Miller D, Clement PB, Crane C, Madore J, Provencher D, Leung P, DeFazio A, Khattra J, Turashvili G, Zhao Y, Zeng T, Glover JN, Vanderhyden B, Zhao C, Parkinson CA, Jimenez-Linan M, Bowtell DD, Mes-Masson AM, Brenton JD, Aparicio SA, Boyd N, Hirst M, Gilks CB, Marra M, Huntsman DG. Mutation of FOXL2 in granulosa-cell tumors of the ovary. N Engl J Med. 360: 2719-2729, 2009.
  235. Lima JF, Jin L, de Araujo AR, Erikson-Johnson MR, Oliveira AM, Sebo TJ, Keeney GL, Medeiros F. FOXL2 mutations in granulosa cell tumors occurring in males. Arch Pathol Lab Med. 136(7):825-828, 2012.
  236. Morrish DW, Venner PM, Siy O, Barron G, Bhardwaj D, Outhet D. Mechanisms of endocrine dysfunction in patients with testicular cancer. J Natl Cancer Inst 82: 412-418, 1990.
  237. van Casteren NJ, Boellaard WP, Romijn JC, Dohle GR. Gonadal dysfunction in male cancer patients before cytotoxic treatment. Int J Androl. 33:c73-79, 2010.
  238. Rives N, Perdrix A, Hennebicq S, Saïas-Magnan J, Melin MC, Berthaut I, Barthélémy C, Daudin M, Szerman E, Bresson JL, Brugnon F, Bujan L. The semen quality of 1158 men with testicular cancer at the time of cryopreservation: results of the French National CECOS Network. J Androl. 33: 1394-1401, 2012.
  239. Haugnes HS, Bosl GJ, Boer H, Gietema JA, Brydøy M, Oldenburg J, Dahl AA, Bremnes RM, Fosså SD. Long-term and late effects of germ cell testicular cancer treatment and implications for follow-up. J Clin Oncol. 30: 3752-3763, 2012.
  240. Fung C, Fossa SD, Williams A, Travis LB. Long-term Morbidity of Testicular Cancer Treatment. Urol Clin North Am. 2015; 42(3):393-408.
  241. Brydøy M, Fosså SD, Klepp O, Bremnes RM, Wist EA, Bjøro T, Wentzel-Larsen T, Dahl O; Norwegian Urology Cancer Group (NUCG) III study group. Sperm counts and endocrinological markers of spermatogenesis in long-term survivors of testicular cancer. Br J Cancer. 107: 1833-1839, 2012.
  242. Hsiao W, Stahl PJ, Osterberg EC, Nejat E, Palermo GD, Rosenwaks Z, Schlegel PN. Successful treatment of postchemotherapy azoospermia with microsurgical testicular sperm extraction: the Weill Cornell experience. J Clin Oncol 29: 1607-1611, 2011.
  243. Gat I, Toren A, Hourvitz A, Raviv G, Band G, Baum M, Lerner-Geva L, Inbar R, Madgar I. Sperm preservation by electroejaculation in adolescent cancer patients. Pediatr Blood Cancer 61:286-90, 2014.
  244. Goossens E, Jahnukainen K, Mitchell RT, van Pelt A, Pennings G, Rives N, Poels J, Wyns C, Lane S, Rodriguez-Wallberg KA, Rives A, Valli-Pulaski H, Steimer S, Kliesch S, Braye A, Andres MM, Medrano J, Ramos L, Kristensen SG, Andersen CY, Bjarnason R, Orwig KE, Neuhaus N, Stukenborg JB.. Fertility preservation in boys: recent developments and new insights. Hum Reprod Open. 2020; 2020(3):hoaa016. doi: 10.1093/hropen/hoaa016.
  245. Diao L, Turek PJ, John CM, Fang F, Reijo Pera RA. Roles of Spermatogonial Stem Cells in Spermatogenesis and Fertility Restoration. Front Endocrinol (Lausanne). 2022 May 12;13:895528. doi: 10.3389/fendo.2022.895528. PMID: 35634498.
  246. Cvancarova M, Samuelsen SO, Magelssen H, Fossa SD. Reproduction rates after cancer treatment: experience from the Norwegian radium hospital. J Clin Oncol 27, 334-343, 2009.
  247. Bandak M, Jørgensen N, Juul A, Vogelius IR, Lauritsen J, Kier MG, Mortensen MS, Glovinski P, Daugaard G. Testosterone deficiency in testicular cancer survivors - a systematic review and meta-analysis. Andrology 4(3):382-388, 2016.
  248. Bandak M, Jørgensen N, Juul A, Lauritsen J, Oturai PS, Mortensen J, Hojman P, Helge JW, Daugaard G. Leydig cell dysfunction, systemic inflammation and metabolic syndrome in long-term testicular cancer survivors. Eur J Cancer 84: 9-17, 2017.
  249. Willemse PM, Burggraaf J, Hamdy NA, Weijl NI, Vossen CY, van Wulften L, van Steijn-van Tol AQ, Rosendaal FR, Osanto S. Prevalence of the metabolic syndrome and cardiovascular disease risk in chemotherapy-treated testicular germ cell tumour survivors. Brit J Cancer 109, 60-67, 2013.
  250. Chovanec M, Abu Zaid M, Hanna N, El-Kouri N, Einhorn LH, Albany C. Long-term toxicity of cisplatin in germ-cell tumor survivors. Ann Oncol 28, 2670-2679, 2017.
  251. Lauritsen J, Hansen MK, Bandak M, Kreiberg MB, Skøtt JW, Wagner T, Gundgaard Kier MG, Holm NV, Agerbæk M, Gupta R, Dehlendorff C, Andersen KK, Daugaard G. Cardiovascular Risk Factors and Disease After Male Germ Cell Cancer. J Clin Oncol 38:584-592, 2020.
  252. Chovanec M, Lauritsen J, Bandak M, Oing C, Kier GG, Kreiberg M, Rosenvilde J, Wagner T, Bokemeyer C, Daugaard G. Late adverse effects and quality of life in survivors of testicular germ cell tumour. Nat Rev Urol. 18(4):227-245, 2021. PMID: 33686290.
  253. Kvammen Ø, Myklebust TÅ, Solberg A, Møller B, Klepp OH, Fosså SD, Tandstad T. Causes of inferior relative survival after testicular germ cell tumor diagnosed 1953-2015: A population-based prospective cohort study. PLoS One. 2019; 14(12):e0225942. doi: 10.1371/journal.pone.0225942.
  254. Hellesnes R, Myklebust TÅ, Fosså SD, Bremnes RM, Karlsdottir Á, Kvammen Ø, Tandstad T, Wilsgaard T, Negaard HFS, Haugnes HS. Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort. J Clin Oncol 39(32):3561-3573, 2021. PMID: 34388002
  255. Milano MT, Dinh PC, Yang H, Zaid MA, Fossa SD, Feldman DR, Monahan PO, Travis LB, Fung C. Solid and Hematologic Neoplasms After Testicular Cancer: A US Population-Based Study of 24 900 Survivors. JNCI Cancer Spectr. 2020 Mar 17;4(3):pkaa017. doi: 10.1093/jncics/pkaa017. PMID: 32455335.
  256. Nichols CR, Nappi L, Kollmannsberger C, Hamilton R, Daneshmand S. Back to the Future-Moving Forward for Testicular Cancer Survivors. JNCI Cancer Spectr. 2019, 4(2):pkz082. doi: 10.1093/jncics/pkz082. PMID: 32190816.
  257. Dahl AA, Haaland CF, Mykletun A, Bremnes R, Dahl O, Klepp O, Wist E, Fosså SD. Study of anxiety disorder and depression in long-term survivors of testicular cancer. J Clin Oncol 23(10):2389-95, 2005. PMID: 15800331.
  258. Skaali T, Fossa SD, Andersson S, Langberg CW, Lehne G, Dahl AA. Is psychological distress in men recently diagnosed with testicular cancer associated with their neuropsychological test performance? Psycho-oncology, 20, 369-377, 2011.
  259. Smith AB, Butow P, Olver I, Luckett T, Grimison P, Toner GC, Stockler MR, Hovey E, Stubbs J, Turner S, Hruby G, Gurney H, Alam M, Cox K, King MT. The prevalence, severity, and correlates of psychological distress and impaired health-related quality of life following treatment for testicular cancer: a survivorship study. J Cancer Surviv 10, 223-233, 2016.
  260. Kreiberg M, Bandak M, Lauritsen J, Andersen KK, Skøtt JW, Johansen C, Agerbaek M, Holm NV, Lau CJ, Daugaard G. Psychological stress in long-term testicular cancer survivors: a Danish nationwide cohort study. J Cancer Surviv. 14(1):72-79, 2020. PMID: 31748852.

 

 

 

 

 

 

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