Summary
Obesity is associated with hypothalamic–pituitary–testicular axis dysregulation in males. Here, we summarize recent evidence derived from clinical trials and studies in preclinical animal models regarding the role of androgen receptor (AR) signaling in the pathophysiology of males with obesity. We also discuss therapeutic strategies targeting the AR for the treatment of obesity and their limitations and provide insight into the future research necessary to advance this field.
Keywords: fat, men, obesity, testosterone
Abbreviations
- AI
aromatase inhibitor
- APs
adipocyte precursors
- AR
androgen receptor
- ARKO
androgen receptor knock out
- BMI
body mass index
- BMPCs
bone marrow precursor cells
- CHH
congenital hypogonadotropic hypogonadism
- DHT
dihydrotestosterone
- E2
estradiol
- EGFR
epidermal growth factor receptor
- ER
estrogen receptor
- ESR
estrogen receptor gene
- FSH
follicle stimulating hormone
- GPCR
g protein coupled receptor
- HFD
high fat diet
- HPT
hypothalamic–pituitary–testes
- LH
luteinizing hormone
- MAP
mitogen associated protein
- ORX
orchidectomy
- PCOS
polycystic ovary syndrome
- SARMS
selective androgen receptor modulators
- SHBG
sex hormone binding globulin
- Tfm
Testicular feminized mouse
1. INTRODUCTION
Obesity represents a major global public health problem with a rapidly growing incidence rate that confers a great socioeconomic burden on our society. According to the World Health Organization (WHO), obesity is characterized by abnormal or excessive fat accumulation that presents a health risk. 1 While the WHO defines obesity as a body mass index (BMI) over 30 kg/m2, this categorical definition does not capture the fact that the risks imparted by increasing body mass follow a continuum and vary considerably between individuals and ethnic groups. 2 In addition, BMI alone is not linearly predictive of mortality 3 and does not capture important aspects of body composition such as distribution of body fat (e.g., visceral adiposity), ectopic fat deposition (e.g., hepatic steatosis), and sarcopenia all of which may modulate cardiometabolic risks in ways not captured by BMI. Importantly, obesity is best understood as a chronic disease of energy imbalance driven by multiple not fully understood pathogenetic mediators (e.g., genetics and dysregulation of satiety factors) driving excess deposition of adipose tissue. The pathologic impact of excess adiposity depends not only on the amount, but also on its distribution and function. Excess adiposity is in turn is associated with multisystem comorbidities (e.g., diabetes, 4 cardiovascular disease, 5 , 6 obstructive sleep apnea, 7 osteoarthritis, 8 increased risk of certain cancers, 9 in some people, depression, 10 and low self esteem 11 ) that confer excess morbidity and mortality, and socioeconomic ramifications. 3 In this context, the American Association of Clinical Endocrinology and the European Association for the Study of Obesity have recently proposed “Adiposity‐Based Chronic Disease” as a clinically more appropriate term for obesity. 12
The Organization for Economic Co‐operation and Development (OECD) reported in over 37 member countries that one in five adults have obesity. 13 This high prevalence translates to an enormous economic burden, with the directs costs in Germany alone estimated to be 29.39 billion euros and indirect costs to be a further 33.65 billion euros. 14 Hormonal mediators such as ghrelin and leptin have been well researched in the pathogenesis of obesity, 15 but this has not led to effective therapy. In contrast, the contribution of sex steroids is less clear. While it is well established that testosterone negatively regulates fat mass in males, the mechanisms by which this hormone exerts its fat‐reducing effects are not well understood. In this review we discuss the evidence from clinical and pre‐clinical studies supporting a role for testosterone action in reducing fat mass, both directly via the androgen receptor (AR), as well as following conversion to dihydrotestosterone (DHT) by the enzyme 5‐alpha reductase, and through the estrogen receptors α and β (ER) via conversion of testosterone to estradiol (E2) by the enzyme aromatase.
The biologic actions of sex steroids may have sex‐specific aspects, 16 and these differing functions may, at least in part, play a role in the sexual dimorphism of body fat distribution. 17 , 18 In men, circulating androgens have been reported to be negatively associated with body fat mass, and in particular with intra‐abdominal fat mass. 19 , 20 In contrast, in women, although the role of androgens in regulating fat mass is less clear, the evidence suggests that androgens are positively associated with fat mass. Studies have reported that bioavailable testosterone is associated with adiposity in peri‐menopausal women. 21 , 22 Moreover, women with polycystic ovary syndrome (PCOS) often exhibit hyperandrogenism which is correlated with abdominal obesity. 23 , 24 Transgender males undergoing gender affirming hormone therapy with testosterone, exhibited a loss in fat mass in android, gynoid, leg, and arm regions. 25 A study in postmenopausal women with obesity administered nandrolone decanoate with concomitant calorie restriction for 9 months reported a reduction in total fat mass compared with patients treated with spironolactone or placebo. 26 However, it is important to note that the loss of fat mass was only seen in subcutaneous fat while there was an increase in visceral fat. 26 Overall, these data are suggestive of a possible dose‐dependent effect of testosterone whereby increasing serum testosterone into the male reference range (either by treatment of hypogonadal men or transgender men) is associated with reductions in fat mass in both sexes. In contrast, intermediate testosterone concentrations, that is, below the male reference range but above the female reference range (e.g., untreated hypogonadal men and women with PCOS) are associated with accumulation of fat in both sexes. 27 Within this review, we will focus on the role of testosterone in regulating fat mass in males. Furthermore, we discuss the amenability of testosterone signaling via the AR as a therapeutic target to reduce fat mass and the research avenues required to generate novel treatments to target this pathway.
2. THE ROLE OF SEX STEROID SIGNALING IN REGULATING FAT MASS—A DISCUSSION OF THE CURRENT EVIDENCE
2.1. Androgen signaling mechanisms
Androgen signaling is complex due to the several endogenous ligands of the AR and numerous molecular pathways by which androgen mediated signaling can occur. Within this review, we focus on the large body of evidence derived from studies investigating the actions of testosterone and DHT; however, it is important to note that there are several other androgens that can exert important biological effects either by direct interaction with the AR or by serving as substrates for local metabolism into testosterone, including 4‐androstendione, 28 11‐ketotestosterone, 29 and DHEAS. 30 , 31 The primary receptor for testosterone and DHT, the AR, is a nuclear transcription factor which can activate both the DNA‐binding‐dependent (genomic) and non‐DNA binding‐dependent (non‐genomic) signaling pathways. Both mechanisms are reviewed in depth elsewhere by ourselves 32 and others. 33 , 34 , 35 In the circulation, testosterone is plasma‐protein bound, tightly to sex hormone binding globulin (SHBG) and loosely to albumin, with only a small percentage circulating as free testosterone. According to the free hormone hypothesis which remains controversial, 36 , 37 free testosterone is the primary bioactive hormone. However, there is evidence that SHBG bound testosterone may have biologic activity potentially via a SHBG membrane receptor. This is discussed in depth in Section 2.6. DNA‐binding‐dependent AR signaling occurs when testosterone diffuses through the cell membrane and binds to the AR, or is converted to the more potent agonist DHT by 5‐alpha reductase, which also binds to the AR. The androgen/AR complex translocates to the nucleus where it binds to androgen response elements to initiate or repress the transcription of target genes. Testosterone can also be aromatized to E2 by the enzyme aromatase which exerts its actions via the ERs by a similar DNA‐binding‐dependent mechanism. This transcriptional regulation of the AR may be further modified by coregulators, molecules that can bind to nuclear transcription factors to magnify or reduce the transcriptional activity. 38 Coregulators of the AR are reviewed extensively elsewhere 39 , 40 , 41 and as such are not included in Figure 1. Testosterone can also elicit cellular changes via rapid signaling of non‐DNA dependent‐pathways which distinguishes them from the DNA‐binding‐dependent pathways. There are numerous non‐DNA binding pathways such as the SHBG receptor 47 and membrane localized calcium channels 48 that have been studied in different tissue types including reproductive, cardiac and skeletal muscles 33 , 34 with some examples depicted in Figure 1. The role of these non‐DNA binding dependent pathways in regulating fat mass, however, has not yet been investigated.
FIGURE 1.

Summary of putative testosterone signaling mechanisms. DNA‐binding dependent signaling (1) occurs via diffusion of testosterone (T) across the cell membrane where it binds directly to the androgen receptor (AR) or is converted by 5α reductase to dihydrotestosterone (DHT), which also binds to the AR. The androgen/AR complex translocates to the nucleus where it binds to androgen response elements to activate or repress the transcription of target genes. T can also be converted to estradiol (E2) by the enzyme aromatase and mediate its actions following binding to estrogen receptors (ER). Examples of non‐DNA binding‐dependent AR signaling pathways are depicted: (2) testosterone activation of ionotropic receptors such as transient receptor potential melastatin 8 (TRPM8) to enable influx of calcium or sodium 35 , 42 , 43 ; (3) testosterone can bind membrane‐bound g protein coupled receptors (GPCR) to activate several canonical GPCR signaling pathways including phospholipase C‐mediated signaling, which regulates calcium levels 44 , 48 ; (4) testosterone activation of membrane‐associated AR which binds to Src and activates the mitogen associated protein (MAP) kinase pathway via transactivation of the epidermal growth factor receptor (EGFR) 45 ; and (5) via non‐EGFR signaling 46
2.2. Obesity and hypogonadism
2.2.1. Clinical evidence of the relationship between hypogonadism and obesity
Male hypogonadism (hypoandrogenism) is a clinical syndrome comprising of clinical symptoms and signs of androgen deficiency in combination with a low serum testosterone concentration. 49 Hypogonadism is classified either as organic (i.e., due to anatomical disease of the hypothalamic–pituitary–testes [HPT] axis), or as functional (due to suppression of the HPT axis by extragonadal disease). 50 Organic hypogonadism is either primary (due to testicular pathology, e.g., Klinefelter's syndrome) or secondary (due to hypothalamic–pituitary disease, e.g., pituitary tumor). Primary hypogonadism is caused by dysfunction of the testes and is associated with low testosterone and high levels of luteinizing hormone (LH) and follicle stimulating hormone (FSH), while secondary hypogonadism is due to dysfunction of the hypothalamus and/or the pituitary gland and is associated with low testosterone and low to (inappropriately) normal levels of LH and FSH. 50 , 51 Organic hypogonadism is due to a specific pathology causing dysfunction of the HPT axis, is usually irreversible and requires life‐long testosterone replacement (unless fertility is desired). Organic hypogonadism can be congenital (e.g., Klinefelter's syndrome or congenital hypogonadotropic hypogonadism [CHH]) or acquired (e.g., testicular injury or pituitary mass lesion).
There are several congenital disorders that result in hypogonadism which exemplify organic hypogonadism. Klinefelter's syndrome is a genetic condition characterized by an extra X chromosome which results in an XXY genotype; however, 20% of cases have additional aneuploidies. 52 Patients with Klinefelter's syndrome present with primary hypogonadism, and it is typically associated with increased body fat mass, particularly abdominal obesity and consequently with an increased risk of metabolic syndrome. 53 The extent to which the metabolic phenotype is caused by testosterone deficiency or by the additional X chromosome is difficult to ascertain. Of note, in CHH which, depending on the genetic defect can be associated with anosmia (i.e., Kallmann's syndrome), an adverse metabolic phenotype is less dominant.
Functional hypogonadism is usually due to hypothalamic–pituitary suppression by extragonadal disease, and obesity is a very common cause. Functional hypogonadism occurs in the absence of recognizable disease of the HPT and can be reversible by treating the disease causing hypothalamic–pituitary suppression, such as weight loss in obesity. 50 , 54 While the definition of hypogonadism comprises both clinical and biochemical evidence of androgen deficiency, in some studies, hypogonadism is defined biochemically. 50 , 54 Of note, clinical features of androgen deficiency are non‐specific, and in a patient with a high burden of comorbidities, it is often difficult to determine whether androgen deficiency‐like clinical features and low testosterone are causally related or whether they merely reflect effects of comorbidities. 55 Obesity is often associated with lowered testosterone and low to normal gonadotrophin concentrations; however, whether this reflects causality, reverse causality, or merely in‐common risk factors remains unclear. 56 , 57 The relationship is likely bidirectional because on the one hand weight loss is associated with an increase in serum testosterone, whereas on the other hand testosterone treatment reduces fat mass. However, it is important to note that, in men with obesity, the effect of substantial weight loss (i.e., generally only achievable only through bariatric surgery) on serum testosterone is arguably more marked than the effect of testosterone treatment on fat mass. 58 , 59 In contrast, the increases in serum testosterone achieved by lifestyle changes are modest around 3 mM, 60 , 61 and difficult to sustain. 62 While a bidirectional Mendelian randomization study suggested that the effect of an elevated BMI on low testosterone is more important than the effect of low testosterone on an increased BMI, such studies require confirmation in independent samples. 63
Effects of obesity on testosterone
Modest obesity is predominantly associated with reduced total testosterone due to insulin resistance associated reductions in its carrier protein sex hormone binding globulin (SHBG). 64 , 65 However, marked obesity (body mass index (BMI) > 35 kg/m2) may lead to genuine suppression of the HPT axis. 56 There are several postulated mechanisms by which obesity may mediate HPT axis suppression including leptin resistance and/or dysregulated insulin signaling. 66 , 67 Additional contributing factors include paternal genetics, 68 leptin resistance, 69 and obesity‐associated elevations in pro‐inflammatory cytokines (reviewed in Grossmann 56 ). Other putative mechanisms such as increased E2 production have been not been confirmed by recent studies using LCMS‐MS based assay technology to accurately measure E2 concentrations in men. 70 , 71
Effects of testosterone treatment on fat mass
Evidence for a role of testosterone in regulating fat mass is provided by numerous studies, of varying quality, that examine the effects of administration of exogenous testosterone on fat mass. 72 , 73 , 74 These studies are heterogenous due to the numerous variables that differ between studies including gonadal state, that is, hypogonadal versus eugonadal, testosterone therapy regimen and selected patient cohorts, thus making comparisons difficult. A systematic review by Huo et al. reporting the effects of testosterone treatment in men with functional hypogonadism 75 showed that of the 25 studies that assessed the effect of testosterone on fat mass, 60% displayed a reduction in fat mass after testosterone treatment. 75 In addition, a meta‐analysis performed by Corona et al. demonstrated a modest −0.34 standardized mean difference in fat mass following testosterone treatment. 59 Subsequently, several additional studies investigating the effects of testosterone on fat mass have been reported and are summarized in Table 1. Together, the majority of studies examining the effect of exogenous testosterone therapy report a modest reduction of fat mass. Isidori et al. reported a ~2 kg reduction of fat mass 83 corroborating evidence from systematic reviews 84 ; however, several questions remain relating to the mechanisms by which testosterone reduces fat mass as well as the long‐term safety of testosterone therapy. Caution, however, must be taken when interpreting these data as a number of confounding factors exist including differences in baseline BMI and gonadal status of the patient cohorts as well as different formulations of testosterone treatment administered. While no rigorous head‐to‐head trials exist, it is plausible that the biologic actions imparted by different formulations of testosterone may differ. For example, it has been suggested that intramuscular testosterone injections are more effective than transdermal gels in increasing lean body weight. 85 Whether this is because intramuscular injection, which eliminates compliance and skin absorption issues, achieve higher testosterone doses than topical (or oral) preparations, and/or whether other factors (such as metabolism, tissue distribution) play a role, has not been fully clarified.
TABLE 1.
Summary of studies post‐2016 demonstrating the effect of testosterone to reduce fat mass
| Brief synopsis | Clinical gonadal state of men in study and method of T measurement | Effect of T treatment on fat mass | Reference |
|---|---|---|---|
| DB, PC, RCT. Men with type 2 diabetes aged 30 to 65 years with HH or eugonadal states were treated with 250 mg of testosterone cypionate intramuscularly every 2 weeks for 23 weeks. Of the 94 men recruited, 50 were eugonadal and 44 had HH. Men with HH were allocated equally to T treatment and placebo. | Fasting testosterone was measured initially and 2 weeks after the first visit. HH was defined as a calculated free T level of <6.5 ng/dl. T measured by LCMS. | Decrease in trunk subcutaneous fat mass (−3.3 kg) but not visceral or hepatic fat mass. | 76 |
| DB, PC, RCT. Men aged 50–70 years with T2D and low T were given testosterone gel (50 mg T daily) increasing to 100 mg T if T levels did not increase, for 24 weeks. | Men had bioavailable T < 7.3 nM. T measured by LCMS. | Average loss of total fat free mass −1.2 kg measured by DEXA. | 77 |
| DB, PC, RCT. Men aged 18–70 with BMI ≥ 30 kg/m2, were given a low caloric diet for 10 weeks followed by 46 weeks of weight maintenance and were supplemented with either Testosterone undecanoate (Injections of 1000 mg at 0, 6, 16, 26, 36, and 46 weeks) or placebo. | Men had a total T level ≤12 nM. T measured by LCMS. | After initial weight loss, men on T therapy maintained weight loss and at study end had lost more fat (−2.9 kg) than their placebo counterparts. However, in a follow up study, loss of fat free mass was not sustained 82 weeks after treatment. | 60 , 62 |
| DB, PC, RCT. Effect of testosterone on atherosclerosis in 308 community dwelling men ≥60 years age was assessed. Patients were randomized and given placebo or 7.5 g of 1% testosterone gel treatment daily for 3 years. Testosterone was titrated to 10 g if serum testosterone was <17.3 nM or 5 g if testosterone was >31.2 nM. | Total T of 3.47 nM to 13.88 nM. Free T of <173.5 pM considered to be low or low‐normal T levels. Immunoassay measurement of T validated against LCMS. | Fat mass was assessed by DEXA. Both groups exhibited increased fat mass, but fat accumulation was significantly less in the T treated group. | 78 |
| DB, PC, RCT. 13 men with Klinefelter's syndrome (av. aged 22–56 years, BMI average 26.7 kg/m2) were given 160 mg (2 doses 80 mg) testosterone undecanoate per day (orally) or placebo for 6 months and compared with 13 age and BMI matched controls. | Gonadal state is unclear as most Klinefelter's patients are supplemented with T. T assessed by LCMS. | Visceral fat mass, total abdominal and intra‐abdominal fat increased while T decreased total body fat and subcutaneous fat mass. | 79 |
| Men with opioid‐induced hypogonadism were randomly assigned to testosterone undecanoate 1000 mg or placebo (injection). | Total bioavailable T average = 2.9 nM. Fasting T levels assessed by LCMS. | T decreased DXA measured total fat mass by 1.2 kg. | 80 |
| 12‐month double‐blinded, placebo‐controlled trial. 101 men <70 years old with cirrhosis and low testosterone were given either testosterone undecanoate (1000 mg intramuscularly) or placebo at 0, 6, 18, 30, and 42 weeks. | Low T was defined as <12 nM measured by immunoassay or Vermeulen calculated free testosterone <230 pM from 2 separate samples. | Patient body composition was assessed by DXA at baseline 6 months and 12 months, patients treated with T exhibited −4.34 kg reduced fat mass. Total lean mass increased by +4.74 kg | 81 |
| Registry‐based, observational study of 823 men average age 60.6 years with a baseline serum total testosterone concentration ≤12.1 nM were treated with testosterone undecanoate (n = 474) every 12 weeks (after an initial 6 weeks booster injection) or untreated (n = 395) and were assessed 2 times a year for 11 years. | All men exhibited symptoms of hypogonadism. Of the 823 trial participants, 474 were obese, 286 overweight, and 63 normal weight. | All patients given testosterone exhibited significant weight loss (normal weight −4.8%, overweight −9.6% and obese −20.6% of body weight), and favorable changes in lipid profiles | 82 |
| DB, PC, RCT, multi‐center trial. 1007 men aged 50–74 years with an average BMI of 34.7 were randomly assignment to placebo or testosterone undecanoate treatment (1000 mg intramuscularly) at baseline, 6 weeks and subsequently every 3 months for 2 years. | Patients were stratified into low (<8.0 nM), medium (8.0 to <11.0 nM), or high (≥11.0 nM) T levels. Average T at baseline was 13.9 nM (placebo) and 13.4 nM (T group). Fasting T levels were measured by validated LCMS assay. | T treatment resulted in a reduction of total fat mass (−2.71 kg) and abdominal fat mass (−2.34%) assessed by DXA. |
Abbreviations: DB, double blind; DEXA, dual‐energy X‐ray absorptiometry; DHT, dihydrotestosterone; HH, hypogonadotropic hypogonadism; LCMS, liquid chromatography and mass spectrometry; PC, placebo controlled; RCT, randomized clinical trial; T, testosterone.
Whether the reduction in fat mass following testosterone treatment in men is depot specific remains unclear, with some studies reporting reductions occurring in subcutaneous 76 , 79 and others, in visceral fat depots. 60 , 86 The specific fat depot in which reductions are noted following testosterone treatment is an important consideration as only increases in intramuscular 87 and visceral fat 88 , 89 as well as an increase in visceral to subcutaneous fat ratio 90 have been associated with increased mortality in men. 91 In contrast, subcutaneous fat mass, especially if located in the gluteofemoral region is associated with decreased all‐cause mortality. 90 However, subcutaneous fat, especially if located in the abdominal region, may not be innocuous. Of note, testosterone has been shown to downregulate lipoprotein lipase in abdominal subcutaneous tissue, which stimulates release of free fatty acids contributing to systemic insulin resistance. 92 In a controlled study, testosterone deprivation increased in insulin resistance despite the lack of an increase in VAT. In this study the testosterone deprivation‐associated increase in insulin resistance was associated with the increase in total fat mass (but not with the decrease in lean mass) collectively suggesting that fat depots other than VAT modulate insulin resistance and that testosterone exerts insulin reducing effects that are, at least in part, independent of changes in VAT.
Reductions in fat mass are among the most consistent effects observed in clinical studies of testosterone treatment in men; however, the amount of fat mass lost differs between studies. This is due to study heterogeneity, with studies including patient with different baseline characteristics (e.g., differing baseline BMI and gonadal states) and differences with respect to treatment length and testosterone formulations used, hence potentially diluting the effect of testosterone treatment. In studies using intramuscular testosterone undecanoate, reductions in fat mass ranged from 3.4 kg 81 to 4.2 kg, 93 In the largest study to date, T4DM, testosterone undecanoate treatment over 2 years, reduced fat mass by 2.7 kg, over and above the effects of a lifestyle program. 74 Skinner et al. reported that intramuscular testosterone injections were more effective than transdermal gels at increasing lean body weight. 85 RCTS to date have been relatively short in duration, with the majority observing testosterone effects over 12 months or less. Observational studies in contrast, 82 , 94 with longer follow up, suggest that testosterone treatment‐associated fat lass is progressive over time with greater amount of fat loss observed in studies with longer duration (Table 1). Moreover, Haider et al. reported in a prospective registry study over 11 years in 178 men with lowered serum testosterone, that testosterone treatment was associated with type 2 diabetes remission in 34.3% of patients. 95 Collectively, these studies suggest that testosterone‐related reductions in fat mass may translate into metabolic benefits. However, it is likely that effects of testosterone treatment on other tissues such as muscle or potentially pancreas may contribute to the metabolic benefits of testosterone. 74
In summary, the clinical evidence from RCTs shows that testosterone treatment given relatively short term (3 months – 24 months) leads to modest reductions in fat mass with two studies reporting a reduction in the risk of type 2 diabetes. In the T4DM trial, testosterone treatment in high risk men over 2 years, reduced the risk of diabetes relative to placebo by 40%. Furthermore, Haider et al. reported in a prospective registry study over 11 years of 178 men with hypogonadism, that 34.3% of patients administered testosterone resulted in remission of their type 2 diabetes 95 suggesting that at least in part, the testosterone‐related reductions in fat mass may translate into metabolic benefits, however, the mechanism by which this occurs remains to be elucidated and is likely not exclusively ascribed to the loss of fat mass. 74 While one of the most consistent effects of testosterone treatment in men is reduction in fat mass and an increase in muscle mass, changes expected to be metabolically favorable, current testosterone treatment guidelines slightly differ in their recommendation regarding indications for testosterone treatment. While all guidelines agree that men with true hypogonadism should (unless they seek fertility) be offered testosterone treatment, metabolic benefits are highlighted in some 96 but not in all guidelines. 49 , 97 While long term benefits and potential risks of testosterone treatment, especially with respect to cardiovascular outcomes, remain unknown. Of note, an ongoing large RCT, the TRAVERSE trial, seeks to evaluate the effect of testosterone therapy on the risk of major adverse cardiovascular events (ClinicalTrials.gov Identifier: NCT03518034). Until more definitive evidence is available, the only unequivocal clinical indication for testosterone treatment is testosterone replacement for men with organic hypogonadism. 49 , 98
2.2.2. Preclinical orchidectomy models to investigate testosterone actions on fat mass
To mimic hypogonadism in men, castration or orchidectomy (ORX) in rodents has been widely utilized. It is well established that ORX in rodents leads to decreased lean body mass 99 , 100 , 101 , 102 , 103 ; however, there are some discrepancies with regard to changes in fat mass. The majority of studies report an increase in whole body adiposity or in specific adipose depots post‐ORX 99 , 104 , 105 , 106 , 107 , 108 ; however, a handful of studies either report no change 101 , 102 or, in one case, a reduction. 103 These discrepancies may be due to different ages of the animals studied as well as the differing length of the studies. 102 Together, the data suggest that ORX and subsequent dysregulation of fat mass may be attributed to reductions in testosterone. The mechanism by which this increase of fat mass occurs due to the reduction of testosterone is unclear; however, recent studies do give evidence to possible mechanisms. Baik et al. reported that castrated mice fed a normal diet exhibited increased adiposity and expression of fatty acid synthesis associated genes. Additionally, concomitant castration and a high fat diet (HFD) resulted in increased expression of the fatty acid transporter CD36 in subcutaneous fat. 104 Similarly, Sebo et al. reported that castrated mice fed an HFD have large adipocytes that are reduced in size by testosterone administration. 106 These studies suggest that loss of testosterone may regulate fatty acid uptake or storage; however, other potential mechanisms by which testosterone regulates fat mass are described below.
The relationship between testosterone and fat mass is further informed by studies that treat orchidectomized animals with testosterone. Sebo et al. reported that testosterone treatment in male mice reduces ORX and HFD associated weight gain with decreases observed in visceral and subcutaneous fat mass as well as adipocyte size. 106 The abrogation of weight gain in orchidectomized rodents via testosterone administration has also been reported by others 100 , 109 with these pre‐clinical data being consistent with the clinical evidence that testosterone treatment reduces fat mass in men.
The mechanisms by which testosterone regulates fat mass remain unclear. Data obtained from in vitro experiments show that testosterone and DHT can inhibit the differentiation of human and BM‐mesenchymal pluripotent cells into adipocytes via AR dependent pathway. 110 , 111 , 112 , 113 While these data provide evidence for a role of testosterone in the inhibition of adipogenesis in pre‐adipocytes, they do not explain the action of testosterone to reduce the mass of pre‐existing fat depots in humans and rodents. Other mechanisms by which testosterone may regulate fat mass include modulation of lipid metabolism, 114 , 115 alteration of muscle metabolism 116 and alteration of mitochondrial function 117 ; with these actions being extensively reviewed elsewhere 118 but further validation is required. Studies in utilizing clinical antagonists that target AR signaling pathways and genetically modified mice have provided further insight into the mechanisms of testosterone action to decrease fat mass.
2.3. The androgen receptor (AR)
2.3.1. Clinical effects of AR inhibition on fat mass
There are few clinical examples of exclusive AR inhibition to draw from when examining the evidence for AR‐mediated regulation of fat mass. One such example is the use of non‐steroidal anti‐androgen (NSAA) compounds such as bicalutamide or flutamide as a monotherapy in prostate cancer patients. Although these compounds inhibit prostate cancer growth by inhibiting androgen binding to the AR, NSAA monotherapy is no longer recommended in favor of androgen deprivation therapy (ADT), which utilize GnRH agonists or antagonists, to abrogate production of endogenous androgens.
In a 12 month open label trial, Smith et al. reported that patients treated with bicalutamide or leuprolide (a GnRH agonist) monotherapies exhibited an increase in whole body fat mass of 11.2% for leuprolide and 6.4% for bicalutamide. 119 Similar increases in fat mass following bicalutamide 120 and enzalutamide 121 , 122 monotherapy have also been reported supporting a role for testosterone acting via the AR to regulate fat mass. It is important to note however, that treatment with AR antagonists also disrupts the negative feedback of testosterone on the central HPT axis, resulting in increased circulating E2, 119 which may also effect body fat. Thus, the increase in fat mass reported in these studies cannot conclusively be ascribed to inhibition of the AR.
2.3.2. The pre‐clinical genetically modified androgen receptor models
The AR is widely expressed in most tissues and cell types making understanding its contribution to physiological processes challenging to unravel. In this review we have broadly grouped these pre‐clinical models into four subtypes. Global AR knockouts (ARKO), where the AR is inactivated in all tissues and cell types (reviewed in Rana et al. 123 ); tissue and/or cell specific ARKOs where the AR is deleted in a specific cell type or tissue while it is expressed normally in all other tissues, 124 AR over‐expression transgenic models where the AR is overexpressed in a specific cell/tissue type 125 and AR gene replacement models in which the AR is expressed in a specific cell‐type while it is inactivated in all other tissues. 126
Testicular feminized mouse (Tfm) model
The Tfm mouse was one of the first rodent models of AR insufficiency. Generated in 1970, 127 these mice possess a single base deletion in the AR resulting in a frameshift mutation and a non‐functional AR protein. 128 Tfm mice have reduced endogenous testosterone levels 129 and develop hepatic steatosis as well as aortic fatty streaks when fed a HFD. 130 , 131 , 132 More recently, Sebo et al. utilized Tfm mice to investigate whether the fat reducing effect of androgens is mediated via an adipocyte‐specific AR dependent mechanism. Following feeding a HFD, Tfm mice displayed an increase in the proliferation of adipocyte precursors (APs) in the subcutaneous but not visceral fat depot compared with control mice as determined by bromodeoxyuridine labelling. This suggests an important role for the AR in regulating subcutaneous fat deposition. However, transplantation of mTomato labelled APs from Tfm mice into subcutaneous fat depots of WT mice fed a HFD did not proliferate at a faster rate compared with WT APs expressing a functional AR, 106 inferring that adipocyte proliferation is not mediated via the AR expressed in APs but may be mediated by another AR dependent mechanism. It is important to note, that the Tfm mouse model has a number of limitations, including potential residual AR activity due to the production of a truncated AR protein. 133 , 134 , 135 The discovery of the Cre/loxP system has enabled the generation of alternative Global‐ARKO mouse models as well as tissue and/or cell specific knockout models of the AR thereby overcoming the limitations of the Tfm mice and have proved advantageous for investigating the mechanism by which testosterone decreases fat mass. 133
Global AR knockout mice
There are several ARKO mouse models generated using the Cre/loxP system which have been reviewed extensively by ourselves 32 , 123 and others 124 , 133 , 136 and are summarized in Table 2. The adipose tissue phenotype of these ARKO mice differs with regards to both the age of onset and fat depots affected. An obese phenotype of increased subcutaneous and visceral fat mass as well as increased numbers of larger adipocytes have been reported in ARKOs fed a regular chow diet from 9–12 weeks of age. 137 , 144 , 147 In contrast, late‐onset obesity characterized by increases in epididymal and perirenal fat pad size as well as increased adipocyte size has been observed at 40 weeks of age. 138 In contrast to the majority of ARKO models which exhibit spontaneous obesity, 143 there is a report of an ARKO model which only displays an obese phenotype when challenged with a HFD. 100 The differences in the fat phenotype of these ARKO models may be attributed to the differences in the genetic background of the mice 126 as well as the region of the AR targeted for deletion which results in either retainment 145 or inactivation 139 , 140 , 143 of the non‐DNA‐dependent actions of the AR in addition to its DNA‐binding‐ dependent actions. 148
TABLE 2.
Summary of the fat phenotype of male global‐ARKO models generated using Cre‐loxP technology
| Targeted AR region and Cre mouse line | Androgen receptor mutation | Fat phenotype | Reference |
|---|---|---|---|
| Exon 1 CMV‐Cre | Recombination results in excision of Exon 1 resulting in a frame shift mutation and no AR protein. | Late onset obesity (12 weeks) with increases in total body adiposity in subcutaneous, infrarenal and intraperitoneal depots but not gonadal. | 137 , 138 , 139 |
| Exon 2 Beta actin (ACTB) ‐Cre | Excision of Exon 2 results in a frameshift mutation and two premature stop codons leading to nonsense mediate decay of mRNA transcript | Late‐onset increased fat accumulation in gonadal and perirenal fat pads, increased adipocyte size at 35 weeks. | 124 , 140 , 141 , 142 |
| Exon 2 phosphoglycerate kinase 1 (PGK1)‐Cre | Deletion of exon 2 results in a similar frameshift mutation to Yeh et al. 140 | Increased adiposity following feeding a HFD characterized by white adipocyte hypertrophy and increased weight of perigonadal and subcutaneous fat pads. | 100 , 143 |
| Exon 3 CMV‐Cre | In‐frame deletion of exon 3 which encodes the second zinc finger of the DNA binding domain (DBD). DNA‐binding‐dependent actions of the AR are abolished, while non‐DNA binding activity remains. | Increased adiposity, specifically increased visceral and subcutaneous fat pads mass and adipocyte hypertrophy at 12 weeks | 144 , 145 |
| Exon 2 CAGGCre‐ER | Excision of exon 2 of the AR results an AR null allele following treatment with tamoxifen. Generated by breeding the AR floxed mouse generated by De Gendt et al. 143 tamoxifen‐inducible CAGGCre‐ER mice. | Pre‐pubertal inactivation of AR results in increased total fat mass and retroperitoneal fat mass. | 146 |
More recently, findings from a tamoxifen‐inducible Global‐ARKO model suggests that the obesity phenotype observed in the previously published Global‐ARKO mouse models is attributed to the loss of AR action pre‐pubertally as post‐pubertal knockdown of the AR had no effect on fat mass. 146 These data, however, must be interpreted with some caution as AR gene knockdown is not complete (approximately 20% mRNA expression remained) and the induction of knockdown via tamoxifen, despite the short induction time (4 days), may pose some effect of fat mass through ER signaling. 149 Together, these data support an action of testosterone via the AR to regulate fat mass. In order to further elucidate the target tissues and/or cells responsible for mediating this effect, tissue‐specific AR knockout and overexpression mouse models have been generated.
2.3.3. Examples of tissue specific androgen receptor knockout models
To determine whether the action of testosterone to decrease fat mass is mediated directly via the AR in adipocytes, several adipocyte‐ARKOs have been generated. Yu et al. and McInnes et al. have both generated adipocyte‐specific ARKOs by crossing floxed AR mice with mice expressing Cre recombinase under the control of the adipocyte protein 2 (aP2). Yu et al. reported male aP2‐ARKOs exhibited no difference in adiposity or morphology of epididymal white adipose tissue at 20 weeks of age when fed a chow diet. 141 Similarly, aP2‐ARKO male mice generated by McInnes et al. displayed no difference in adipose weight of the subcutaneous or mesenteric regions and a statistically significant but biologically small reduction in perigonadal fat mass at 12 weeks of age compared with chow fed wild‐type controls. No difference in adipocyte area was observed in either the perigonadal or subcutaneous fat depots. Despite this lack of difference in fat pad mass in aP2‐ARKOs fed a standard chow diet, following challenging the mice with a HFD for 24 weeks, mesenteric and omental fat pad mass were increased in aP2‐ARKOs compared with wild‐type controls. 150 The lack of spontaneous obesity in ap2‐ARKO mice suggests that the action of testosterone to decrease fat mass is mediated via the AR in another target tissue. It is important to note that the aP2 promoter is also expressed in several cell types in the brain which may confound these findings as AR action in the brain may indirectly regulate fat mass by regulating voluntary activity. 151
Evidence from muscle‐specific ARKOs suggest that the AR in myocytes may contribute to the fat reducing effects of testosterone. Myocyte‐specific deletion of the AR using mice expressing Cre recombinase under the control of the muscle creatine kinase promoter (MCK‐Cre) resulted in decreased intra‐abdominal fat and lean body mass. 152 In contrast, deletion of the AR specifically in satellite cells using the myoblast determination protein 1 (MyoD‐iCre) promoter mice had no effect on fat mass as measured by DXA. 153 This discrepancy may be due to the different activities of the Cre promoters as the improved Cre with increased Cre expression was used to generate the MyoD‐Cre mice. Irrespective, the action of AR via muscle in the regulation of fat mass remains unclear.
2.3.4. AR expression in bone marrow cells
More recently, bone has been recognized for its importance as an endocrine organ that may regulate numerous metabolic functions through the secretion of osteokines. Once such example is osteocalcin which can regulate several physiological functions including glucose metabolism, 154 energy expenditure, 155 and testosterone levels. 156 Investigation into the potential role of androgen action via the AR in bone marrow cells in negatively regulating fat mass is an exciting research direction, with current papers related to this topic summarized in Table 3.
TABLE 3.
Reports pertaining to the effects of AR signaling in bone marrow cells on adiposity
| Model | AR expressing cell | Fat and metabolic phenotype | Reference |
|---|---|---|---|
| BM transplant (ARKO) | WT or ARKO bone marrow was transplanted into WT mice and fed HFD for 16 weeks. | Visceral fat mass was increased in WT mice that received ARKO bone marrow 8 weeks post‐HCD and BM transplantation. No effect was observed at 16 weeks post‐HCD and BM transplantation. | 157 |
| 3.6‐kb α1 1 collagen promoter AR over expression (Col3.6 AR transgenic) | AR overexpression in bone marrow progenitor cells, and their descendants (i.e., osteoblasts and osteocytes) | DHT treatment of Col3.6 AR transgenics reverses ORX‐mediated adiposity. | 158 , 159 |
| 2.3‐kb type 1α1 collagen promoter AR over expression (Col2.3 AR transgenic) | AR overexpression in mature and mineralizing osteoblasts and osteocytes. | No difference in adiposity. | 125 , 141 |
| PC‐AR Gene Replacement mice | Expression of the AR in BMPCsCol3.6 while it is deleted in all other tissues. Generated by breeding Global‐ARKO mice 145 with Col3.6 AR transgenics 159 | Expression of the AR in BMPCs on a global ARKO background results in complete attenuation of the increased fat mass of global ARKOs. | 126 |
| Monocyte/macrophage specific ARKO | Deletion of the AR in monocytes and macrophages. Generated by breeding floxed AR and Lyz2‐Cre 160 mice. | No effect on adiposity. | 160 |
| AR‐ null BMPCs | Bone marrow progenitor cells isolated from Global‐ARKOs 161 | Loss of the AR in BMPCs promoted adipogenesis and inhibited osteogenesis. | 162 |
Utilizing a unique bone marrow transplant model whereby the bone marrow of ARKO mice was grafted into irradiated C57BL/6 J WT mice, Rubinow et al. demonstrated that WT mice receiving ARKO bone marrow exhibited significantly higher visceral and total fat mass than WT mice receiving WT bone marrow when fed a HFD diet. 157 In a subsequent study, the authors narrowed their focus to the specific bone marrow monocyte and macrophage cell type due to their endogenous expression of AR as well as their documented role in energy metabolism. 160 Deletion of the AR specifically in monocytes/macrophages expressing the lysozyme 2 gene did not affect adiposity. 160 Intriguingly, while the loss of the AR in bone marrow cells confers a propensity to obesity, this is not mediated by lysozyme 2 expressing monocytes/macrophages, suggesting that another cell type in the bone marrow may be responsible for mediating the fat reducing effects of testosterone.
Bone marrow precursor cells (BMPCs) are pluripotent cells that can differentiate into osteoprogenitors, osteoblasts, osteocytes, chondrocytes and adipocytes. 163 Given the significant contribution of BMPCs to the various cellular components of the bone and the evidence that bone may act as endocrine organ, 164 , 165 it stands to reason that these BMPCs may play a role in the regulation of fat mass. One of the first studies investigating the action of the AR via BMPCs utilized the 3.6 kb α1 1 collagen promoter to overexpress the AR in BMPCs (Col3.6AR‐Transgenics [Tg]). Male Col3.6AR‐Tg mice displayed reduced visceral fat mass in the gonadal and peri‐renal fat depots and smaller adipocyte area in the absence of any changes in serum adipokines. 166 Of significant interest, DHT treatment was able to reverse the increase in fat accumulation that occurs following gonadectomy in both adult male and female Col3.6AR‐Tgs but not WT mice. 158 In contrast, overexpression of the AR in mature osteoblasts of male mice utilizing the 2.3‐kb α1(1) collagen promoter (Col2.3AR‐Tg) had no effect on fat mass 125 indicating that the decrease in fat mass is mediated at an earlier stage of osteoblast development, that is, in BMPCs and osteoprogenitors.
Recently, we provided evidence for an action of androgens via the AR in bone marrow BMPCs to negatively regulate fat mass and improve metabolic function. 126 Replacement of the AR gene specifically in BMPCs of our Global‐ARKOs to generate PC‐AR Gene Replacement mice, completely attenuates their increased fat mass. 126 PC‐AR gene replacement mice had an increase in the number of smaller adipocytes and a healthier metabolic profile, characterized by normal serum leptin, elevated serum adiponectin and improved whole‐body insulin sensitivity, with higher glucose uptake into subcutaneous and visceral fat than Global‐ARKOs. 126
Collectively, these studies utilizing genetically modified mouse models provide evidence to support the notion that the action of testosterone to negatively regulate fat mass, is at least in part, mediated directly via the AR. While the current data do not support the adipocyte or myocyte as the target cells for this AR action, BMPCs appear to be a strong candidate for further study.
While our discussion on the metabolic syndrome is focused on AR action, it is important to note that, in men, the presence of the metabolic syndrome is associated with many factors, including but not limited to age, marital status, recent paternity, and lifestyle factors (e.g., sedentary behavior). Of note in men, such factors are also associated with circulating testosterone concentrations, 167 hence intertwining the metabolic syndrome and serum testosterone in complex ways, making the disentanglement of pathogenic links between circulating testosterone and the metabolic syndrome a complex endeavor.
2.4. 5 α reductase and dihydrotestosterone (DHT)
5 α reductase (5αR) is the enzyme which converts testosterone to DHT, a more potent, non aromatizable, androgen that regulates the formation of the external male genitalia and the prostate. 168 5αR and DHT are integral to AR signaling but clinical data suggest that 5αR and DHT are not strongly associated with the regulation of fat mass. This is evidenced by the absence of weight gain in patients with prostate cancer treated with the 5αR inhibitor, finasteride. 169 Moreover, in a mechanistic RCT in healthy men, the fat‐reducing effects of testosterone treatment were not altered by co‐administration of the 5αR inhibitor dutasteride, implying that the testosterone‐mediated reduction in fat mass does not require its reduction to DHT with the caveat that the RCT may not have been sufficiently powered to make this conclusion. 170 , 171 In a separate study, Juang et al. abrogated endogenous sex steroid production through the GnRH agonist acycline, in healthy men and randomized them to three treatment groups or a fourth placebo group. The three treatment arms were testosterone add‐back only, testosterone add‐back combined with dutasteride (to suppress DHT), and testosterone add‐back combined with the aromatase inhibitor (AI) anastrozole (to suppress E2). As expected, patients treated with testosterone exhibited significant reductions in fat mass. Inhibition of aromatase abrogated these effects but consistent with the previous findings of Bhasin et al., 170 5αR inhibition did not alter the effects of testosterone. 172 Taken together, this evidence suggests that while DHT does not play role in the fat reducing effects of testosterone, the fat reducing effects of testosterone may require its aromatization to E2 (see Section 2.5). Conversely, some evidence points to a role for DHT in regulating fat mass. 173 Idan et al. reported in a randomized, placebo controlled trial of 114 older men that DHT treatment over 2 years increased lean mass but decreased fat mass, akin to the effects of testosterone. 174 Of note, DHT treatment was associated with reduced circulating E2, implying that there was no confounding effect of increased E2 on fat mass, but rather, the effects were directly related to DHT.
Overall, the evidence suggests that 5αR and by this extension, DHT, is less significant in regulating fat mass than testosterone, and that the fat reducing effects of testosterone may not be exclusively mediated through the AR. One possible alternative mechanism inferred by the work of Juang and Finkelstein could be, at least in men, the conversion of testosterone to E2 (see Section 2.5).
2.4.1. 5 α reductase KO mice and alternative murine models
There are a handful of studies investigating the role of 5αR and DHT in the regulation of adipose tissue in murine models with largely incongruent results. Sato et al. report a reduction of subcutaneous fat mass in male mice treated with DHT. 139 More recently, Sebo et al. demonstrated a decrease in subcutaneous fat following DHT treatment in orchidectomized mice fed a HFD, although to a lesser extent than observed with testosterone, 106 and Kim et al. reported no effect of DHT on overall fat mass in HFD fed, chemically castrated mice. 175 There have been also several reports of 5αR type 1 KO mice with no reports of altered fat 176 including in gonadal depots. 177 The only piece of data contradictory to this was reported by Movérare‐Skrtic et al., who reported in male orchidectomized mice treated with DHT for 5 weeks has been shown to increase gonadal and retroperitoneal fat mass. 178 However, interestingly, the authors report no statistically significant changes in retroperitoneal or gonadal fat mass in ORX only mice. Taken together these data suggest that while DHT may be able to negatively modulate fat mass, its contribution is not as marked as testosterone and may be restricted to subcutaneous depots.
2.5. Estradiol, aromatase, and the estrogen receptor
There is evidence from both human and pre‐clinical studies suggesting that the fat reducing effects of testosterone are, at least in part, dependent on its aromatization to E2. As the role of E2 as a male hormone has been reviewed extensively elsewhere, 71 , 179 below we summarize here the key role of testosterone derived E2 in regulating fat mass.
2.5.1. Clinical evidence of the role of estradiol and aromatase mediated reduction of fat mass
An important role for E2 in limiting fat mass in men is supported by observations that men with inactivating mutations in the CYP19 gene which encodes aromatase, and men with inactivating mutations of the ERα have increased BMI and abdominal adipose tissue. 71 , 180 Mechanistic studies in men where testosterone is administrated concomitantly with an AI, 172 , 180 , 181 , 182 , 183 suggest that the fat reducing effects of testosterone are at least in part dependent on its aromatization to E2.
Upon appraisal of the literature as well as data from other reviews, 71 , 179 we conclude that aromatization of testosterone to E2 is required to mediate at least some of the fat reducing effects of testosterone. However, studies using AI have inferred the role of E2 by reducing its serum concentrations, and no study in men to date has assessed the direct effects of E2 in the absence of T.
2.5.2. Preclinical evidence of estradiol and aromatase regulation of fat mass
Preclinical models are useful for dissecting the role of E2 in mediating the actions of testosterone on fat mass. Male aromatase KO mice have increased adiposity compared with controls despite having elevated circulating levels of testosterone due to the lack of negative feedback of E2 on the HPT axis, 106 , 184 , 185 , 186 , 187 analogous to men with aromatase deficiency. In one report, male aromatase KO mice exhibited increased gonadal adipose tissue which was ameliorated by E2 treatment. 188 Consistent with these findings, treatment of intact mice with the aromatase inhibitor, letrozole, was shown to increase fat mass compared with vehicle‐treated mice 106 and the administration of E2 to castrated mice fed a HFD was effective in decreasing diet‐induced weight gain. 106 These data support the notion that in addition to the action of testosterone directly via the AR to decrease fat mass, testosterone can also exert these effects following conversion to E2 and action via the ER. Data generated with cell‐specific overexpression or deficiency of aromatase is somewhat contradictory. Bone marrow transplantation from aromatase deficient mice into irradiated wild type mice results in a small, but statically significant increase in fat mass at 8 weeks post‐transplantation that was no longer evident by 16 weeks. 189 Male mice that overexpress aromatase specifically in white adipose tissue have decreased subcutaneous and gonadal fat mass, but somewhat unexpectedly have increased retroperitoneal fat mass. 190 It is difficult to consolidate the discrepancy between cell‐specific and whole body aromatase knockout data; however, we may speculate, due to the wide spread expression of aromatase, 191 that the specific cells salient to modulating the effect of aromatase on fat mass are yet to be discovered.
2.5.3. Clinical evidence of estrogen receptor mediated signaling regulating fat mass
Estrogen receptors (ER) are expressed broadly in both men and women. 71 , 192 There are two ER subtypes, ER α and ER β, cytosolically located nuclear transcription factor receptors that signal via DNA‐binding‐dependent signaling pathways. A third, more recently characterized G‐protein coupled ER, which is localized in the membrane and signals via canonical G protein coupled receptor signaling pathways, provides another avenue of ER mediated signaling. 193 The role of estrogens in regulating body fat distribution has been reviewed in detail elsewhere 194 , 195 but of specific interest to this review is the direct contribution of ER signaling in testosterone‐mediated fat loss.
To date, only two men with mutations in the estrogen receptor gene (ESR) 1 which encodes ER α have been reported. One patient had a BMI in the obese range of 30 kg/m2 and clinical evidence of insulin resistance, 196 although fat mass was not reported. This phenotype resembles that of men with inactivating mutations in the aromatase gene. 180 The second patient had a BMI of 23.7 kg/m2, but also had low serum testosterone due to cryptorchidism. 197 Overall, the data from these rare case reports are consistent with the notion that the lifelong absence of E2 signaling in men, via ER alpha may limit fat mass accumulation and promote insulin resistance. Of note, men with inactivating mutations in the ESR 2 gene encoding ER β have not been, to our knowledge, reported.
There are currently no definitive mechanistic studies that precisely quantify the contribution of ER signaling in testosterone‐mediated fat loss. This may be in part attributed to the lack of appropriate pharmacological compounds available that specifically bind one of the three ER subtypes. In an optimal situation, complete co‐administration of a GnRH agonist (to achieve confounding suppression of endogenous testicular sex steroid production) and of an ER receptor specific antagonist would allow the dissection of the role of specific ER receptors in the regulation of fat mass. However, some mechanistic studies have been performed that yield some evidence for a role of estradiol signaling in regulating fat mass by utilizing two classes of compounds; aromatase inhibitors and Selective Estrogen Receptor Modulators (SERMS). Aromatase inhibitors such as anastrozole have been reported to increase fat loss when combined with weight loss programs in men affected by obesity 198 and in a RCT of older men with hypogonadism. 199 Of note, these findings which imply that inhibition of E2 promotes fat loss are contrary to Finkelstein's work 181 reporting that E2 is required to mediate the fat reducing action of testosterone. These differences could be partially explained by the differences in age and baseline body composition of the participants in the three trials. Compounds such as clomiphene and tamoxifen are SERMS and act as mixed agonist‐antagonists of the ER depending on the tissue specific expression and subtype specific expression of the ER. 200 It is important to note however, that SERMs and aromatase inhibitors, via diminishing estradiol mediated negative feedback on GnRH and gonadotrophin secretion, can increase serum testosterone concentrations thereby confounding interpretation of data. 201 The few studies investigating the use of clomiphene citrate to treat adult men with overweight/obesity, have reported no 202 to moderate 203 reductions in fat mass. Thus, the clinical evidence to date does not provide a conclusive picture on the role of E2 in the regulation of fat mass. Evidence attempting to decipher the role of ER signaling in testosterone mediated fat reductions has also arisen from pre‐clinical models.
2.5.4. Pre‐clinical evidence of estrogen receptor mediated signaling regulating fat mass
There are numerous animal models that modulate the ER signaling pathway. These are predominantly receptor KO models and have provided insight into the role of ER signaling in the regulation of fat mass. Male ERα KO mice have increased fat mass 204 , 205 , 206 , 207 ; while in contrast, ER β KO mice exhibit no fat phenotype, 207 , 208 , 209 suggesting that ERα but not ER β regulates fat mass. However, it is important to note, that there is one report of adipocyte‐specific knock down of the ERα using siRNA that had no effect on fat mass in male mice. 210 Several additional cell specific ERα KO mice have been generated including osteocytes 211 and hepatocytes 212 that show no change in fat phenotype.
Despite the absence of any reported changes in fat deposition in global ERβ KO mice, agonistic ERβ‐selective ligands have been reported to mitigate diet‐induced obesity in male mice. 213 Activation of ERβ using 4‐(2‐(3‐5‐dimethylisoxazol‐4‐yl)‐1H‐indol‐3yl) phenol (DIP) can alter fat deposition in male mice to a female deposition. 214 In addition, strong interactions between E2 and leptin signaling have been reported in male rats 179 indicating that ER signaling has the potential to also modulate fat mass through indirect mechanisms such as decreasing satiety. In summary, these data suggest that activation of ERα may play a role in the regulation of fat mass in males, but further study is required to ascertain the contribution of the fat reducing effect ERβ activation and the mechanisms by which it exerts these effects. Estradiol has been reported to regulate lipid and glucose metabolism 210 , 215 , 216 representing potential mechanisms.
2.6. Sex hormone binding globulin (SHBG)
Testosterone is present in plasma in a free form and a bound form. Bound testosterone is predominantly bound to the sex hormone binding globulin (SHBG) and albumin. SHBG binds testosterone and DHT with high affinity and E2 with a comparatively lower affinity. 217 Of note, SHBG is inversely associated in men with obesity, 64 , 218 however, whether SHBG contributes to the pathogenesis of obesity in men is not known.
Androgen binding protein (ABP) is synthesized by Sertoli cells of the testis in rodents and humans and has a similar amino acid structure to SHBG, but differs in carbohydrate composition. 219 While ABP has been shown to regulate the accumulation and bioavailability of testosterone within the seminiferous tubular epithelium, 220 , 221 there is, to our knowledge, no evidence of association of ABP with obesity or fat mass.
2.6.1. Clinical evidence of SHBG contribution to testosterone, and regulated fat reduction
Obesity has been reported to be associated with lower circulating levels of SHBG. 64 , 222 SHBG is increased by resistance training with concomitant increases in lean body mass and reductions in total and trunk fat mass. 223 Additionally, bariatric surgery has been reported to reverse reduced testosterone and SHBG concentrations in men with obesity. 224 In congruence with this, the reduction of visceral and subcutaneous fat mass through the use of lifestyle intervention and metformin has been reported to increase SHBG levels. 225 This infers there is an association between SHBG and obesity, but whether this is causal, due to reverse causality or merely reflects in‐common risk factors is not known. Pre‐clinical models have provided more evidence in this regard.
2.6.2. Pre‐clinical evidence of a role for SHBGs in testosterone‐mediated fat reduction
As hepatic SHBG is not expressed in rodent models, 226 pre‐clinical rodent models have utilized genetic expression of human SHBG 227 in order to study its role in mediating the action of testosterone to regulate fat mass. One such example is a mouse model that over‐expresses human SHBG on the db/db mouse model of obesity (leptin KO model). db/db mice over‐expressing SHBG were less obese than control db/db mice expressing normal levels of SHBG 228 consistent with observations in humans 64 indicating that SHBG is inversely associated with obesity. This same model has been used to demonstrate the importance of SHBG in regulating sex steroid activity 226 and interestingly, may protect against HFD induced obesity through the induction of lipolysis in white adipose tissue, 229 though this finding is contradicted elsewhere. 230 Given these contradictory findings combined with the use of over‐expression models with supraphysiological expression of SHBG, it is not possible to ascertain whether SHBG has a direct role in the regulation of fat mass, an indirect role via modulation of circulating sex steroid concentrations, or whether changes in circulating SHBG are simply a marker of adiposity.
3. AR‐REGULATED PATHWAYS MAY BE AMENABLE FOR THERAPEUTIC TARGETING—LIMITATIONS AND FUTURE DIRECTIONS
Obesity is a debilitating disease with numerous co‐morbidities that confers a significant socioeconomic burden. Obesity's wide, growing prevalence and recalcitrance to treatment makes finding more effective treatments of immediate importance. There are several novel treatments being developed that are reviewed elsewhere 231 and exhibit promise; however, it is of benefit to generate a suite of anti‐obesity compounds that may enable specific obesity therapies. Testosterone has been studied as a potential therapy for hypogonadism related obesity and has been reported to have moderate efficacy. 74
Understanding how molecular signaling pathways lead to physiological or pathophysiological changes has long been a key way of identifying therapies for diseases including cancer 232 , 233 and diabetes. Alternative therapies that signal through the AR pathways to reduce fat mass without masculinizing side effects are a promising therapeutic avenue. 234
3.1. Current limitations to using the AR as a therapeutic target for obesity
There are several limitations that prevent targeting the AR for obesity. These can be broadly grouped into two categories: (1) understanding of the mechanisms of fat loss and (2) generation of specific targeting compounds.
3.1.1. Understanding AR signaling mechanisms to reduce fat mass
A mechanistic understanding of how androgens regulate body composition has long been sought after to enable more efficacious therapies. 166 Current evidence suggests that the fat sparing effects of testosterone may be mediated through a variety of cells other than adipocytes, 150 including muscle cells. 152 , 153 , 235 In addition, more recent evidence discussed in this review suggests that androgen action via the AR in BMPCs decreases fat mass, 126 , 166 , 189 is an exciting avenue of research.
3.1.2. Undiscovered osteokines, and adipokines released from the bone
One of the unresolved questions is whether AR signaling modulates body composition via direct effects, that is, tissue specific (autocrine or paracrine) AR signaling, and/or via indirect endocrine effects, that is, via stimulating the secretion of yet to be identified circulating effectors. It is well understood that the bone, 165 muscle 236 and adipose tissues 237 secrete numerous cytokines, some of which represent an unexamined source of potential novel therapeutic targets. Additionally BMPCs have also been reported to secrete factors and regulate metabolic function both in a paracrine and endocrine capacity. 238 , 239 Testosterone has been reported to regulate the secretion of metabolic factors such as adiponectin 240 and leptin. 241 Therefore, it is conceivable that that the AR may regulate the secretion of novel yet to be discovered factors, which in turn may decrease fat mass. Thus, it is possible that some of these novel, or yet to be discovered secreted factors may form the basis of new treatments.
This notion of novel inter‐cellular communication molecules forming the basis of novel therapeutics have been previously reported. 242 Hormones were identified and subsequently leveraged for use as therapeutics throughout the 20th century but it is only in more recent times that numerous families of secreted signaling molecules bearing the suffix “‐kines” such as myokines, 243 osteokines, adipokines, 244 and hepatokines 245 have been identified. 242 The first identified adipokine that was thought to have value as a therapeutic was leptin, 246 but leptin therapy alone has failed as a therapy for obesity. Since then, numerous other signaling molecules have been identified but have yet to yield results, 242 suggesting there may be other opportunities that remain to be discovered.
3.1.3. Compounds that leverage unique AR signaling mechanisms
The AR signaling pathway elicits its biological effects through both DNA‐binding‐dependent and non‐DNA‐binding‐dependent pathways 32 which may be activated by either cytosolically located AR or membrane localized AR 46 as well as other membrane localized receptors. 35 This offers opportunities to generate specific novel compounds that activate desired pathways. One such example of this was the recently developed testosterone loaded monosialoganglioside micelles, which were able to bypass the effects of membrane‐initiated signaling to target DNA binding‐dependent signaling exclusively by the micelle directly delivering testosterone to the cytoplasm. This represents an exciting avenue for enabling novel therapeutics 247 but also requires a clearer understanding of the contribution of DNA binding‐ and non‐DNA binding‐ dependent AR signaling to the regulation of fat mass.
A class of compounds termed selective androgen receptor modulators (SARMs) 248 present another potential opportunity to specifically activate the AR pathways responsible for regulating fat mass. A better understanding of the mechanisms by which SARMs exhibit tissue selectivity would greatly benefit the development of novel compounds. Putative mechanisms of SARM selectivity include tissue specific enzymes, modulated recruitment of coregulators and activation of unique subcellular signaling cascades are some of the putative mechanisms of SARM selectivity, 249 but more research is required to understand these mechanisms and then leverage these unique features to generative more efficacious therapeutics. Generating SARMs that exclusively target the AR mediated mechanisms of fat reduction may hold an alternative approach to treating obesity. However, SARMs have a number of potential limitations. Considering the work of Finkelstein et al. 181 demonstrating the importance of the aromatization of testosterone to estradiol and ER signaling in the reduction of testosterone‐mediated fat mass, SARMs may be less potent in fat mass reduction compared with testosterone due to their non‐aromatizable nature. Furthermore, the utilization of SARMs as therapeutics opens several important questions regarding the effect of SARMs on physiological parameters such as bone health and fertility. However, as highlighted in this review, there are important knowledge gaps with regards to the mechanism by which androgens regulate fat mass and more research is needed in order to inform the utility of targeting the AR signaling pathway as a treatment for obesity.
4. CONCLUSION
It is apparent that a two‐pronged approach is required to enable the targeting of unique AR signaling pathways, that is understanding the signaling mechanisms that traditional AR agonists such as testosterone and DHT utilize to reduce fat mass or elicit other positive effects, as well as developing novel compounds that can target these molecular processes with minimal negative side effects.
Obesity is a multifactorial disease 250 that requires novel therapies in conjunction with current lifestyle and pharmacological interventions in order to be effectively treated. In this review we have discussed the importance of AR signaling in the regulation of fat mass and highlight the lack of research into this molecular pathway. By investigating AR signaling pathways that reduce fat mass and identifying novel therapeutic targets to leverage this pathway, we may ultimately be able to offer new treatments for this debilitating disease and reduce the immense burden placed on society. This review has focused on males, but such novel therapies may also be of value to females affected by obesity. However, as highlighted in this review, there are important knowledge gaps with regards to the mechanism by which androgens regulate fat mass and more research is needed in order to inform the utility of targeting the AR signaling pathway as a treatment for obesity.
CONFLICT OF INTEREST
Dr. Grossmann reports grants and personal fees from Bayer Healthcare and personal fees from Besins Healthcare, outside the submitted work. Drs. Davey, Venkatesh, and Zajac have nothing to disclose.
ACKNOWLEDGEMENTS
Open access publishing facilitated by The University of Melbourne, as part of the Wiley ‐ The University of Melbourne agreement via the Council of Australian University Librarians.
Venkatesh VS, Grossmann M, Zajac JD, Davey RA. The role of the androgen receptor in the pathogenesis of obesity and its utility as a target for obesity treatments. Obesity Reviews. 2022;23(6):e13429. doi: 10.1111/obr.13429
REFERENCES
- 1. World Health Organisation . World Health Organisation: obesity and overweight fact sheet. https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight. Published 2021. Accessed December 3, 2021.
- 2. Di Angelantonio E, Bhupathiraju SN, Wormser D, et al. Body‐mass index and all‐cause mortality: individual‐participant‐data meta‐analysis of 239 prospective studies in four continents. Lancet. 2016;388(10046):776‐786. doi: 10.1016/S0140-6736(16)30175-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Tobias DK, Hu FB. The association between BMI and mortality: implications for obesity prevention. Lancet Diabetes Endocrinol. 2018;6(12):916‐917. doi: 10.1016/S2213-8587(18)30309-7 [DOI] [PubMed] [Google Scholar]
- 4. Okamura T, Hashimoto Y, Hamaguchi M, Obora A, Kojima T, Fukui M. Ectopic fat obesity presents the greatest risk for incident type 2 diabetes: a population‐based longitudinal study. Int J Obes (Lond). 2019;43(1):139‐148. doi: 10.1038/s41366-018-0076-3 [DOI] [PubMed] [Google Scholar]
- 5. Berrington de Gonzalez A, Hartge P, Cerhan JR, et al. Body‐mass index and mortality among 1.46 million White adults. N Engl J Med. 2010;363(23):2211‐2219. doi: 10.1056/nejmoa1000367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Twig G, Yaniv G, Levine H, et al. Body‐mass index in 2.3 million adolescents and cardiovascular death in adulthood. N Engl J Med. 2016;374(25):2430‐2440. doi: 10.1056/nejmoa1503840 [DOI] [PubMed] [Google Scholar]
- 7. Kuna ST, Reboussin DM, Borradaile KE, et al. Long‐term effect of weight loss on obstructive sleep apnea severity in obese patients with type 2 diabetes. Sleep. 2013;36(5):641‐649. doi: 10.5665/sleep.2618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bliddal H, Leeds AR, Christensen R. Osteoarthritis, obesity and weight loss: evidence, hypotheses and horizons—a scoping review. Obes Rev. 2014;15(7):578‐586. doi: 10.1111/obr.12173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. De Pergola G, Silvestris F. Obesity as a major risk factor for cancer. J Obes. 2013;2013;291546. doi: 10.1155/2013/291546 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bartolome F, Antequera D, Tavares E, et al. Obesity and neuroinflammatory phenotype in mice lacking endothelial megalin. J Neuroinflammation. 2017;14(1):1‐10. doi: 10.1186/s12974-017-0800-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Luppino FS, de Wit LM, Bouvy PF, et al. Overweight, obesity, and depression: a systematic review and meta‐analysis of longitudinal studies. Arch Gen Psychiatry. 2010;67(3):220‐229. doi: 10.1001/archgenpsychiatry.2010.2 [DOI] [PubMed] [Google Scholar]
- 12. Timothy GW. New horizons. A new paradigm for treating to target with second‐generation obesity medications. J Clin Endocrinol Metab. 2021;XX(Xx):1‐9. doi: 10.1210/clinem/dgab848 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. OECD . Obesity update 2017. Diabetologe. 2017;13(5):331‐341. doi: 10.1007/s11428-017-0241-7 [DOI] [Google Scholar]
- 14. Effertz T, Engel S, Verheyen F, Linder R. The costs and consequences of obesity in Germany: a new approach from a prevalence and life‐cycle perspective. Eur J Health Econ. 2016;17(9):1141‐1158. doi: 10.1007/s10198-015-0751-4 [DOI] [PubMed] [Google Scholar]
- 15. Oussaada SM, van Galen KA, Cooiman MI, et al. The pathogenesis of obesity. Metabolism. 2019;92:26‐36. doi: 10.1016/j.metabol.2018.12.012 [DOI] [PubMed] [Google Scholar]
- 16. Schiffer L, Kempegowda P, Arlt W, O'Reilly MW. Mechanisms in endocrinology: the sexually dimorphic role of androgens in human metabolic disease. Eur J Endocrinol. 2017;177(3):R125‐R143. doi: 10.1530/EJE-17-0124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Blouin K, Boivin A, Tchernof A. Androgens and body fat distribution. J Steroid Biochem Mol Biol. 2008;108(3–5):272‐280. doi: 10.1016/j.jsbmb.2007.09.001 [DOI] [PubMed] [Google Scholar]
- 18. Bond ST, Calkin AC, Drew BG. Sex differences in white adipose tissue expansion: emerging molecular mechanisms. Clin Sci (Lond). 2021;135(24):2691‐2708. doi: 10.1042/CS20210086 [DOI] [PubMed] [Google Scholar]
- 19. Pasquali R, Casimirri F, Cantobelli S, et al. Effect of obesity and body fat distribution on sex hormones and insulin in men. Metabolism. 1991;40(1):101‐104. doi: 10.1016/0026-0495(91)90199-7 [DOI] [PubMed] [Google Scholar]
- 20. Khaw KT, Barrett‐Connor E. Lower endogenous androgens predict central adiposity in men. Ann Epidemiol. 1992;2(5):675‐682. doi: 10.1016/1047-2797(92)90012-F [DOI] [PubMed] [Google Scholar]
- 21. Janssen I, Powell LH, Jasielec MS, Kazlauskaite R. Covariation of change in bioavailable testosterone and adiposity in midlife women. Obesity. 2015;23(2):488‐494. doi: 10.1002/oby.20974 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kalyani RR, Franco M, Dobs AS, et al. The association of endogenous sex hormones, adiposity, and insulin resistance with incident diabetes in postmenopausal women. J Clin Endocrinol Metab. 2009;94(11):4127‐4135. doi: 10.1210/jc.2009-0910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dumesic DA, Akopians AL, Madrigal VK, et al. Hyperandrogenism accompanies increased intra‐abdominal fat storage in normal weight polycystic ovary syndrome women. J Clin Endocrinol Metab. 2016;101(11):4178‐4188. doi: 10.1210/jc.2016-2586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Polak AM, Adamska A, Krentowska A, et al. Body composition, serum concentrations of androgens and insulin resistance in different polycystic ovary syndrome phenotypes. J Clin Med. 2020;9(3);732. doi: 10.3390/jcm9030732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Klaver M, De Blok CJM, Wiepjes CM, et al. Changes in regional body fat, lean body mass and body shape in trans persons using cross‐sex hormonal therapy: results from a multicenter prospective study. Eur J Endocrinol. 2018;178(2):163‐171. doi: 10.1530/EJE-17-0496 [DOI] [PubMed] [Google Scholar]
- 26. Lovejoy JC, Bray GA, Bourgeois MO, et al. Exogenous androgens influence body composition and regional body fat distribution in obese postmenopausal women—a clinical research center study. J Clin Endocrinol Metab. 1996;81(6):2198‐2203. doi: 10.1210/jcem.81.6.8964851 [DOI] [PubMed] [Google Scholar]
- 27. Schiffer L, Arlt W, O'Reilly MW. Understanding the role of androgen action in female adipose tissue. Front Horm Res. 2019;53:33‐49. doi: 10.1159/000494901 [DOI] [PubMed] [Google Scholar]
- 28. Jasuja R, Ramaraj P, Mac RP, et al. Δ‐4‐androstene‐3,17‐dione binds androgen receptor, promotes myogenesis in vitro, and increases serum testosterone levels, fat‐free mass, and muscle strength in hypogonadal men. J Clin Endocrinol Metab. 2005;90(2):855‐863. doi: 10.1210/jc.2004-1577 [DOI] [PubMed] [Google Scholar]
- 29. Imamichi Y, Yuhki KI, Orisaka M, et al. 11‐Ketotestosterone is a major androgen produced in human gonads. J Clin Endocrinol Metab. 2016;101(10):3582‐3591. doi: 10.1210/jc.2016-2311 [DOI] [PubMed] [Google Scholar]
- 30. Storbeck KH, Bloem LM, Africander D, Schloms L, Swart P, Swart AC. 11β‐hydroxydihydrotestosterone and 11‐ketodihydrotestosterone, novel C19 steroids with androgenic activity: a putative role in castration resistant prostate cancer? Mol Cell Endocrinol. 2013;377(1–2):135‐146. doi: 10.1016/j.mce.2013.07.006 [DOI] [PubMed] [Google Scholar]
- 31. Mo Q, Lu S, fang, Simon NG. Dehydroepiandrosterone and its metabolites: differential effects on androgen receptor trafficking and transcriptional activity. J Steroid Biochem Mol Biol. 2006;99(1):50‐58. doi: 10.1016/j.jsbmb.2005.11.011 [DOI] [PubMed] [Google Scholar]
- 32. Davey RA, Grossmann M. Androgen receptor structure, function and biology: from bench to bedside. Clin Biochem Rev. 2016;37(1):3‐15. doi: 10.1038/sc.1992.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Rahman F, Christian HC. Non‐classical actions of testosterone: an update. Trends Endocrinol Metab. 2007;18(10):371‐378. doi: 10.1016/j.tem.2007.09.004 [DOI] [PubMed] [Google Scholar]
- 34. Lucas‐Herald AK, Alves‐Lopes R, Montezano AC, Ahmed SF, Touyz RM. Genomic and non‐genomic effects of androgens in the cardiovascular system: clinical implications. Clin Sci. 2017;131(13):1405‐1418. doi: 10.1042/CS20170090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Thomas P. Membrane androgen receptors unrelated to nuclear steroid receptors. Endocrinology. 2019;160(4):772‐781. doi: 10.1210/en.2018-00987 [DOI] [PubMed] [Google Scholar]
- 36. Handelsman DJ. Free testosterone: pumping up the tires or ending the free ride? Endocr Rev. 2017;38(4):297‐301. doi: 10.1210/er.2017-00171 [DOI] [PubMed] [Google Scholar]
- 37. Goldman AL, Bhasin S, Wu FCW, Krishna M, Matsumoto AM, Jasuja R. A reappraisal of testosterone's binding in circulation: physiological and clinical implications. Endocr Rev. 2017;38(4):302‐324. doi: 10.1210/ER.2017-00025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. McKenna NJ, Lanz RB, O'Malley BW. Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev. 1999;20(3):321‐344. doi: 10.1210/er.20.3.321 [DOI] [PubMed] [Google Scholar]
- 39. Heinlein CA, Chang C. Androgen receptor (AR) coregulators: an overview. Endocr Rev. 2002;23(2):175‐200. doi: 10.1210/edrv.23.2.0460 [DOI] [PubMed] [Google Scholar]
- 40. Veilleux A, Blouin K, Tchernof A. Mechanisms of androgenic action in adipose tissue. Future Lipidol. 2009;4(3):367‐378. doi: 10.2217/CLP.09.16 [DOI] [Google Scholar]
- 41. van de Wijngaart DJ, Dubbink HJ, van Royen ME, Trapman J, Jenster G. Androgen receptor coregulators: recruitment via the coactivator binding groove. Mol Cell Endocrinol. 2012;352(1–2):57‐69. doi: 10.1016/j.mce.2011.08.007 [DOI] [PubMed] [Google Scholar]
- 42. Mohandass A, Krishnan V, Gribkova ED, et al. TRPM8 as the rapid testosterone signaling receptor: implications in the regulation of dimorphic sexual and social behaviors. FASEB J. 2020;34(8):10887‐10906. doi: 10.1096/fj.202000794R [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Asuthkar S, Elustondo PA, Demirkhanyan L, et al. The TRPM8 protein is a testosterone receptor. J Biol Chem. 2015;290(5):2659‐2669. doi: 10.1074/jbc.M114.610824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Shihan M, Bulldan A, Scheiner‐Bobis G. Non‐classical testosterone signaling is mediated by a G‐protein‐coupled receptor interacting with Gnα11. Biochim Biophys Acta ‐ Mol Cell Res. 2014;1843(6):1172‐1181. doi: 10.1016/j.bbamcr.2014.03.002 [DOI] [PubMed] [Google Scholar]
- 45. Cheng J, Watkins SC, Walker WH. Testosterone activates mitogen‐activated protein kinase via Src kinase and the epidermal growth factor receptor in sertoli cells. Endocrinology. 2007;148(5):2066‐2074. doi: 10.1210/en.2006-1465 [DOI] [PubMed] [Google Scholar]
- 46. Deng Q, Zhang Z, Wu Y, et al. Non‐genomic action of androgens is mediated by rapid phosphorylation and regulation of androgen receptor trafficking. Cell Physiol Biochem. 2017;43(1):223‐236. doi: 10.1159/000480343 [DOI] [PubMed] [Google Scholar]
- 47. Rosner W, Hryb DJ, Kahn SM, Nakhla AM, Romas NA. Interactions of sex hormone‐binding globulin with target cells. Mol Cell Endocrinol. 2010;316(1):79‐85. doi: 10.1016/j.mce.2009.08.009 [DOI] [PubMed] [Google Scholar]
- 48. Vicencio JM, Ibarra C, Estrada M, et al. Testosterone induces an intracellular calcium increase by a nongenomic mechanism in cultured rat cardiac myocytes. Endocrinology. 2006;147(3):1386‐1395. doi: 10.1210/en.2005-1139 [DOI] [PubMed] [Google Scholar]
- 49. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an endocrine society. J Clin Endocrinol Metab. 2018;103(5):1715‐1744. doi: 10.1210/jc.2018-00229 [DOI] [PubMed] [Google Scholar]
- 50. Grossmann M, Matsumoto AM. A perspective on middle‐aged and older men with functional hypogonadism: focus on holistic management. J Clin Endocrinol Metab. 2017;102(3):1067‐1075. doi: 10.1210/jc.2016-3580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Salonia A, Rastrelli G, Hackett G, et al. Paediatric and adult‐onset male hypogonadism. Nat Rev Dis Primers. 2019;5:38. doi: 10.1038/s41572-019-0087-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Panimolle F, Radicioni AF. 47,XXY Klinefelter syndrome is associated with an increased risk of insulin resistance: the impact of hypogonadism and visceral obesity. Diabetes Assoc with Single Gene Defects Chromosom Abnorm. 2017;25:151‐159. doi: 10.1159/000454742 [DOI] [Google Scholar]
- 53. Bojesen A, Kristensen K, Birkebaek NH, et al. The metabolic syndrome is frequent in Klinefelter's syndrome and is associated with abdominal obesity and hypogonadism. Diabetes Care. 2006;29(7):1591‐1598. doi: 10.2337/dc06-0145 [DOI] [PubMed] [Google Scholar]
- 54. Corona G, Rastrelli G, Morelli A, et al. Treatment of functional hypogonadism besides pharmacological substitution. World J Mens Health. 2019;37(3):256‐270. doi: 10.5534/WJMH.190061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sterling J, Bernie AM, Ramasamy R. Opinion hypogonadism: easy to define, hard to diagnose, and controversial to treat. Canadian Urol Assoc J. 2020;9(February 2015):65‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Grossmann M. Hypogonadism and male obesity: focus on unresolved questions. Clin Endocrinol (Oxf). 2018;89(1):11‐21. doi: 10.1111/cen.13723 [DOI] [PubMed] [Google Scholar]
- 57. Glass AR, Swerdloff RS, Bray GA, Dahms WT, Atkinson RL. Low serum testosterone and sex‐hormone‐binding‐globulin in massively obese men. J Clin Endocrinol Metab. 1977;45(6):1211‐1219. doi: 10.1210/jcem-45-6-1211 [DOI] [PubMed] [Google Scholar]
- 58. Lee Y, Dang JT, Switzer N, et al. Impact of bariatric surgery on male sex hormones and sperm quality: a systematic review and meta‐analysis. Obes Surg. 2019;29(1):334‐346. doi: 10.1007/s11695-018-3557-5 [DOI] [PubMed] [Google Scholar]
- 59. Corona G, Giagulli VA, Maseroli E, et al. Testosterone supplementation and body composition: results from a meta‐analysis of observational studies. J Endocrinol Invest. 2016;39(9):967‐981. doi: 10.1007/s40618-016-0480-2 [DOI] [PubMed] [Google Scholar]
- 60. Ng Tang Fui M, Prendergast LA, Dupuis P, et al. Effects of testosterone treatment on body fat and lean mass in obese men on a hypocaloric diet: a randomised controlled trial. BMC Med. 2016;14:153. doi: 10.1186/s12916-016-0700-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Corona G, Giagulli VA, Maseroli E, et al. Testosterone supplementation and body composition: results from a meta‐analysis study. Eur J Endocrinol. 2016;174(3):R99‐R116. doi: 10.1530/EJE-15-0262 [DOI] [PubMed] [Google Scholar]
- 62. Ng Tang Fui M, Hoermann R, Zajac JD, Grossmann M. The effects of testosterone on body composition in obese men are not sustained after cessation of testosterone treatment. Clin Endocrinol (Oxf). 2017;87(4):336‐343. doi: 10.1111/cen.13385 [DOI] [PubMed] [Google Scholar]
- 63. Eriksson J, Haring R, Grarup N, et al. Causal relationship between obesity and serum testosterone status in men: a bidirectional mendelian randomization analysis. PLoS ONE. 2017;12(4):1‐15. doi: 10.1371/journal.pone.0176277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Cooper LA, Page ST, Amory JK, Anawalt BD, Matsumoto AM. The association of obesity with sex hormone‐binding globulin is stronger than the association with ageing ‐ implications for the interpretation of total testosterone measurements. Clin Endocrinol (Oxf). 2015;83(6):828‐833. doi: 10.1111/cen.12768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Peter A, Kantartzis K, Machann J, et al. Relationships of circulating sex hormone‐binding globulin with metabolic traits in humans. Diabetes. 2010;59(12):3167‐3173. doi: 10.2337/db10-0179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Dhindsa S, Ghanim H, Batra M, Dandona P. Hypogonadotropic hypogonadism in men with diabesity. Diabetes Care. 2018;41(7):1516‐1525. doi: 10.2337/dc17-2510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Grossmann M, Ng Tang Fui M, Cheung AS. Late‐onset hypogonadism: metabolic impact. Andrology. 2020;8(6):1519‐1529. doi: 10.1111/andr.12705 [DOI] [PubMed] [Google Scholar]
- 68. Sanchez‐Garrido MA, Ruiz‐Pino F, Velasco I, et al. Intergenerational influence of paternal obesity on metabolic and reproductive health parameters of the offspring: male‐preferential impact and involvement of kiss1‐mediated pathways. Endocrinology. 2018;159(2):1005‐1018. doi: 10.1210/en.2017-00705 [DOI] [PubMed] [Google Scholar]
- 69. Izquierdo AG, Crujeiras AB, Casanueva FF, Carreira MC. Leptin, obesity, and leptin resistance: where are we 25 years later? Nutrients. 2019;11(11):1‐11. doi: 10.3390/nu11112704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. 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. doi: 10.2337/dc11-0208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Russell N, Grossmann M. Estradiol as a male hormone. Eur J Endocrinol. 2019;181(1):R23‐R43. doi: 10.1530/EJE-18-1000 [DOI] [PubMed] [Google Scholar]
- 72. Frederiksen L, Højlund K, Hougaard DM, et al. Testosterone therapy decreases subcutaneous fat and adiponectin in aging men. Eur J Endocrinol. 2012;166(3):469‐476. doi: 10.1530/EJE-11-0565 [DOI] [PubMed] [Google Scholar]
- 73. Mårin P, Holmtäng S, Gustafsson C, et al. Androgen treatment of abdominally obese men. Obes Res. 1993;1(4):245‐251. doi: 10.1002/j.1550-8528.1993.tb00618.x [DOI] [PubMed] [Google Scholar]
- 74. Wittert G, Bracken K, Robledo KP, et al. Testosterone treatment to prevent or revert type 2 diabetes in men enrolled in a lifestyle programme (T4DM): a randomised, double‐blind, placebo‐controlled, 2‐year, phase 3b trial. Lancet Diabetes Endocrinol. 2021;9(1):32‐45. doi: 10.1016/S2213-8587(20)30367-3 [DOI] [PubMed] [Google Scholar]
- 75. Huo S, Scialli AR, McGarvey S, et al. Treatment of men for “low testosterone”: a systematic review. PLoS ONE. 2016;11(9):1‐46. doi: 10.1371/journal.pone.0162480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Dhindsa S, Ghanim H, Batra M, et al. Insulin resistance and inflammation in hypogonadotropic hypogonadism and their reduction after testosterone replacement in men with type 2 diabetes. Diabetes Care. 2016;39(1):82‐91. doi: 10.2337/dc15-1518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Magnussen LV, Hvid LG, Hermann AP, et al. Testosterone therapy preserves muscle strength and power in aging men with type 2 diabetes—a randomized controlled trial. Andrology. 2017;5(5):946‐953. doi: 10.1111/andr.12396 [DOI] [PubMed] [Google Scholar]
- 78. Huang G, Pencina KM, Li Z, et al. Long‐term testosterone administration on insulin sensitivity in older men with low or low‐normal testosterone levels. J Clin Endocrinol Metab. 2018;103(4):1678‐1685. doi: 10.1210/jc.2017-02545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Høst C, Bojesen A, Erlandsen M, et al. A placebo‐controlled randomized study with testosterone in klinefelter syndrome: beneficial effects on body composition. Endocr Connect. 2019;8(9):1250‐1261. doi: 10.1530/EC-19-0323 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Glintborg D, Vaegter HB, Christensen LL, et al. Testosterone replacement therapy of opioid‐induced male hypogonadism improved body composition but not pain perception: a double‐blind, randomized, and placebo‐controlled trial. Eur J Endocrinol. 2020;182(6):539‐548. doi: 10.1530/EJE-19-0979 [DOI] [PubMed] [Google Scholar]
- 81. Sinclair M, Grossmann M, Hoermann R, Angus PW, Gow PJ. Testosterone therapy increases muscle mass in men with cirrhosis and low testosterone: a randomised controlled trial. J Hepatol. 2016;65(5):906‐913. doi: 10.1016/j.jhep.2016.06.007 [DOI] [PubMed] [Google Scholar]
- 82. Saad F, Doros G, Haider KS, Haider A. Differential effects of 11 years of long‐term injectable testosterone undecanoate therapy on anthropometric and metabolic parameters in hypogonadal men with normal weight, overweight and obesity in comparison with untreated controls: real‐world data from. Int J Obes (Lond). 2020;44(6):1264‐1278. doi: 10.1038/s41366-019-0517-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Isidori AM, Giannetta E, Greco EA, et al. Effects of testosterone on body composition, bone metabolism and serum lipid profile in middle‐aged men: a meta‐analysis. Clin Endocrinol (Oxf). 2005;63(3):280‐293. doi: 10.1111/j.1365-2265.2005.02339.x [DOI] [PubMed] [Google Scholar]
- 84. Corona G, Rastrelli G, Monami M, et al. Body weight loss reverts obesity‐associated hypogonadotropic hypogonadism: a systematic review and meta‐analysis. Eur J Endocrinol. 2013;168(6):829‐843. doi: 10.1530/EJE-12-0955 [DOI] [PubMed] [Google Scholar]
- 85. Skinner JW, Otzel DM, Bowser A, et al. Muscular responses to testosterone replacement vary by administration route: a systematic review and meta‐analysis. J Cachexia Sarcopenia Muscle. 2018;9(3):465‐481. doi: 10.1002/jcsm.12291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Boyanov MA, Boneva Z, Christov VG. Testosterone supplementation in men with type 2 diabetes, visceral obesity and partial androgen deficiency. Aging Male. 2003;6(1):1‐7. doi: 10.1080/tam.6.1.1.7 [DOI] [PubMed] [Google Scholar]
- 87. Koster A, Murphy RA, Eiriksdottir G, et al. Fat distribution and mortality: the AGES‐Reykjavik study. Obesity. 2015;23(4):893‐897. doi: 10.1002/oby.21028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Abraham TM, Pedley A, Massaro JM, Hoffmann U, Fox CS. Association between visceral and subcutaneous adipose depots and incident cardiovascular disease risk factors. Circulation. 2015;132(17):1639‐1647. doi: 10.1161/CIRCULATIONAHA.114.015000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Gruzdeva O, Borodkina D, Uchasova E, Dyleva Y, Barbarash O. Localization of fat depots and cardiovascular risk. Lipids Health Dis. 2018;17:218. doi: 10.1186/s12944-018-0856-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Lee SW, Son JY, Kim JM, Hwang SS, Han JS, Heo NJ. Body fat distribution is more predictive of all‐cause mortality than overall adiposity. Diabetes Obes Metab. 2018;20(1):141‐147. doi: 10.1111/dom.13050 [DOI] [PubMed] [Google Scholar]
- 91. Vasan SK, Osmond C, Canoy D, et al. Comparison of regional fat measurements by dual‐energy X‐ray absorptiometry and conventional anthropometry and their association with markers of diabetes and cardiovascular disease risk. Int J Obes (Lond). 2018;42(4):850‐857. doi: 10.1038/ijo.2017.289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Mårin P, Odén B, Björntorp P. Assimilation and mobilization of triglycerides in subcutaneous abdominal and femoral adipose tissue in vivo in men: effects of androgens. J Clin Endocrinol Metab. 1995;80(1):239‐243. doi: 10.1210/jcem.80.1.7829619 [DOI] [PubMed] [Google Scholar]
- 93. Svartberg J, Agledahl I, Figenschau Y, Sildnes T, Waterloo K, Jorde R. Testosterone treatment in elderly men with subnormal testosterone levels improves body composition and BMD in the hip. Int J Impot Res. 2008;20(4):378‐387. doi: 10.1038/ijir.2008.19 [DOI] [PubMed] [Google Scholar]
- 94. Saad F, Yassin A, Doros G, Haider A. Effects of long‐term treatment with testosterone on weight and waist size in 411 hypogonadal men with obesity classes I‐III: observational data from two registry studies. Int J Obes (Lond). 2016;40(1):162‐170. doi: 10.1038/ijo.2015.139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Haider KS, Haider A, Saad F, et al. Remission of type 2 diabetes following long‐term treatment with injectable testosterone undecanoate in patients with hypogonadism and type 2 diabetes: 11‐year data from a real‐world registry study. Diabetes Obes Metab. 2020;22(11):2055‐2068. doi: 10.1111/dom.14122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Garvey WT, Mechanick JI, Brett EM, et al. American association of clinical endocrinologists and American college of endocrinology comprehensive clinical practice guidelines for medical care of patients with obesity. Endocr Pract. 2016;22(July):1‐203. doi: 10.4158/EP161365.GL [DOI] [PubMed] [Google Scholar]
- 97. Yeap BB, Grossmann M, McLachlan RI, et al. Endocrine Society of Australia position statement on male hypogonadism (Part 2): treatment and therapeutic considerations. Med J Aust. 2016;205(5):228‐231. doi: 10.5694/mja16.00448 [DOI] [PubMed] [Google Scholar]
- 98. Lapauw B, Kaufman JM. Management of endocrine disease: rationale and current evidence for testosterone therapy in the management of obesity and its complications. Eur J Endocrinol. 2020;183(6):R16‐R183. doi: 10.1530/EJE-20-0394 [DOI] [PubMed] [Google Scholar]
- 99. Vandenput L, Boonen S, Van Herck E, Swinnen JV, Bouillon R, Vanderschueren D. Evidence from the aged orchidectomized male rat model that 17β‐estradiol is a more effective bone‐sparing and anabolic agent than 5α‐dihydrotestosterone. J Bone Miner Res. 2002;17(11):2080‐2086. doi: 10.1359/jbmr.2002.17.11.2080 [DOI] [PubMed] [Google Scholar]
- 100. Dubois V, Laurent MR, Jardi F, et al. Androgen deficiency exacerbates high‐fat diet‐induced metabolic alterations in male mice. Endocrinology. 2016;157(2):648‐665. doi: 10.1210/en.2015-1713 [DOI] [PubMed] [Google Scholar]
- 101. Vanderschueren D, Vandenput L, Boonen S, Van Herck E, Swinnen JV, Bouillon R. An aged rat model of partial androgen deficiency: prevention of both loss of bone and lean body mass by low‐dose androgen replacement. Endocrinology. 2000;141(5):1642‐1647. doi: 10.1210/endo.141.5.7472 [DOI] [PubMed] [Google Scholar]
- 102. Moreau MF, Libouban H, Legrand E, Baslé MF, Audran M, Chappard D. Lean, fat and bone masses are influenced by orchidectomy in the rat. A densitometric X‐ray absorptiometric study. J Musculoskelet Neuronal Interact. 2001;1(3):209‐20913. [PubMed] [Google Scholar]
- 103. Floryk D, Kurosaka S, Tanimoto R, et al. Castration‐induced changes in mouse epididymal white adipose tissue. Mol Cell Endocrinol. 2011;345(1–2):58‐67. doi: 10.1016/j.mce.2011.07.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Baik M, Jeong JY, Park SJ, et al. Testosterone deficiency caused by castration increases adiposity in male rats in a tissue‐specific and diet‐dependent manner. Genes Nutr. 2020;15(1):1‐10. doi: 10.1186/s12263-020-00673-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Faust IM, Johnson PR, Kral JG. Effects of castration on adipose tissue growth and regrowth in the male rat. Metabolism. 1984;33(7):596‐601. doi: 10.1016/0026-0495(84)90055-6 [DOI] [PubMed] [Google Scholar]
- 106. Sebo ZL, Rodeheffer MS. Testosterone metabolites differentially regulate obesogenesis and fat distribution. Mol Metab. 2021;44(December 2020):1‐9. doi: 10.1016/j.molmet.2020.101141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Aoki A, Fujitani K, Takagi K, Kimura T, Nagase H, Nakanishi T. Male hypogonadism causes obesity associated with impairment of hepatic gluconeogenesis in mice. Biol Pharm Bull. 2016;39(4):587‐592. doi: 10.1248/bpb.b15-00942 [DOI] [PubMed] [Google Scholar]
- 108. Harada N, Hanaoka R, Horiuchi H, et al. Castration influences intestinal microflora and induces abdominal obesity in high‐fat diet‐fed mice. Sci Rep. 2016;6(October 2015):1‐9. doi: 10.1038/srep23001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Arinno A, Apaijai N, Kaewthep P, et al. Combination of low‐dose testosterone and vildagliptin confers cardioprotection in castrated obese rats. J Endocrinol. 2019;240(3):467‐481. doi: 10.1530/JOE-18-0673 [DOI] [PubMed] [Google Scholar]
- 110. Gupta V, Bhasin S, Guo W, et al. Effects of dihydrotestosterone on differentiation and proliferation of human mesenchymal stem cells and preadipocytes. Mol Cell Endocrinol. 2008;296(1–2):32‐40. doi: 10.1016/j.mce.2008.08.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Blouin K, Nadeau M, Perreault M, et al. Effects of androgens on adipocyte differentiation and adipose tissue explant metabolism in men and women. Clin Endocrinol (Oxf). 2010;72(2):176‐188. doi: 10.1111/j.1365-2265.2009.03645.x [DOI] [PubMed] [Google Scholar]
- 112. Bhasin S, Taylor WE, Singh R, et al. The mechanisms of androgen effects on body composition: mesenchymal pluripotent cell as the target of androgen action. J Gerontol Ser A Biol Sci Med Sci. 2003;58(12):M1103‐M1110. doi: 10.1093/gerona/58.12.m1103 [DOI] [PubMed] [Google Scholar]
- 113. Singh R, Artaza JN, Taylor WE, et al. Testosterone inhibits adipogenic differentiation in 3T3‐L1 cells: nuclear translocation of androgen receptor complex with β‐catenin and T‐cell factor 4 may bypass canonical Wnt signaling to down‐regulate adipogenic transcription factors. Endocrinology. 2006;147(1):141‐154. doi: 10.1210/en.2004-1649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Maseroli E, Comeglio P, Corno C, et al. Testosterone treatment is associated with reduced adipose tissue dysfunction and nonalcoholic fatty liver disease in obese hypogonadal men. J Endocrinol Invest. 2020;44(4):819‐842. doi: 10.1007/s40618-020-01381-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Høst C, Gormsen LC, Christensen B, et al. Independent effects of testosterone on lipid oxidation and VLDL‐TG production: a randomized, double‐blind, placebo‐controlled, crossover study. Diabetes. 2013;62(5):1409‐1416. doi: 10.2337/db12-0440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Sarchielli E, Comeglio P, Filippi S, et al. Testosterone improves muscle fiber asset and exercise performance in a metabolic syndrome model. J Endocrinol. 2020;245(2):259‐279. doi: 10.1530/JOE-19-0532 [DOI] [PubMed] [Google Scholar]
- 117. Usui T, Kajita K, Kajita T, et al. Elevated mitochondrial biogenesis in skeletal muscle is associated with testosterone‐induced body weight loss in male mice. FEBS Lett. 2014;588(10):1935‐1941. doi: 10.1016/j.febslet.2014.03.051 [DOI] [PubMed] [Google Scholar]
- 118. Kelly DM, Jones TH. Testosterone and obesity. Obes Rev. 2015;16(7):581‐606. doi: 10.1111/obr.12282 [DOI] [PubMed] [Google Scholar]
- 119. Smith MR, Goode M, Zietman AL, McGovern FJ, Lee H, Finkelstein JS. Bicalutamide monotherapy versus leuprolide monotherapy for prostate cancer: Effects on bone mineral density and body composition. J Clin Oncol. 2004;22(13):2546‐2553. doi: 10.1200/JCO.2004.01.174 [DOI] [PubMed] [Google Scholar]
- 120. Wadhwa VK, Weston R, Parr NJ. Bicalutamide monotherapy preserves bone mineral density, muscle strength and has significant health‐related quality of life benefits for osteoporotic men with prostate cancer. BJU Int. 2011;107(12):1923‐1929. doi: 10.1111/j.1464-410X.2010.09726.x [DOI] [PubMed] [Google Scholar]
- 121. Tombal B, Borre M, Rathenborg P, et al. Enzalutamide monotherapy in hormone‐naive prostate cancer: primary analysis of an open‐label, single‐arm, phase 2 study. Lancet Oncol. 2014;15(6):592‐600. doi: 10.1016/S1470-2045(14)70129-9 [DOI] [PubMed] [Google Scholar]
- 122. Tombal B, Borre M, Rathenborg P, et al. Long‐term antitumor activity and safety of enzalutamide monotherapy in hormone naïve prostate cancer: 3‐year open label followup results. J Urol. 2018;199(2):459‐464. doi: 10.1016/j.juro.2017.08.103 [DOI] [PubMed] [Google Scholar]
- 123. Rana K, Davey RA, Zajac JD. Human androgen deficiency: insights gained from androgen receptor knockout mouse models. Asian J Androl. 2014;16(2):169‐177. doi: 10.4103/1008-682X.122590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Chang C, Yeh S, Lee SO, Chang TM. Androgen receptor (AR) pathophysiological roles in androgen‐related diseases in skin, bone/muscle, metabolic syndrome and neuron/immune systems: lessons learned from mice lacking AR in specific cells. Nucl Recept Signal. 2013;11:1‐26. doi: 10.1621/nrs.11001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Wiren KM, Semirale AA, Zhang XW, et al. Targeting of androgen receptor in bone reveals a lack of androgen anabolic action and inhibition of osteogenesis. A model for compartment‐specific androgen action in the skeleton. Bone. 2008;43(3):440‐451. doi: 10.1016/j.bone.2008.04.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Russell PK, Mangiafico S, Fam BC, et al. The androgen receptor in bone marrow progenitor cells negatively regulates fat mass. J Endocrinol. 2018;237(1):15‐27. doi: 10.1530/JOE-17-0656 [DOI] [PubMed] [Google Scholar]
- 127. Lyon MF, Hawkes SG. X‐linked gene for testicular feminization in the mouse. Nature. 1970;227(5264):1217‐1219. doi: 10.1038/2271217a0 [DOI] [PubMed] [Google Scholar]
- 128. Charest NJ, Zhou Z, Lubahn DB, Olsen KL, Wilson EM, French FS. A frameshift mutation destabilizes androgen receptor messenger RNA in the Tfm mouse. Mol Endocrinol. 1991;5(4):573‐581. doi: 10.1210/mend-5-4-573 [DOI] [PubMed] [Google Scholar]
- 129. Murphy L, O'Shaughnessy PJ. Testicular steroidogenesis in the testicular feminized (Tfm) mouse: loss of 17α‐hydroxylase activity. J Endocrinol. 1991;131(3):443‐449. doi: 10.1677/joe.0.1310443 [DOI] [PubMed] [Google Scholar]
- 130. Kelly DM, Akhtar S, Sellers DJ, Muraleedharan V, Channer KS, Jones TH. Testosterone differentially regulates targets of lipid and glucose metabolism in liver, muscle and adipose tissues of the testicular feminised mouse. Endocrine. 2016;54(2):504‐515. doi: 10.1007/s12020-016-1019-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kelly DM, Nettleship JE, Akhtar S, et al. Testosterone suppresses the expression of regulatory enzymes of fatty acid synthesis and protects against hepatic steatosis in cholesterol‐fed androgen deficient mice. Life Sci. 2014;109(2):95‐103. doi: 10.1016/j.lfs.2014.06.007 [DOI] [PubMed] [Google Scholar]
- 132. Kelly DM, Sellers DJ, Woodroofe MN, Jones TH, Channer KS. Effect of testosterone on inflammatory markers in the development of early atherogenesis in the testicular‐feminized mouse model. Endocr Res. 2013;38(3):125‐138. doi: 10.3109/07435800.2012.735307 [DOI] [PubMed] [Google Scholar]
- 133. Walters KA, Simanainen U, Handelsman DJ. Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Hum Reprod Update. 2010;16(5):543‐558. doi: 10.1093/humupd/dmq003 [DOI] [PubMed] [Google Scholar]
- 134. He WW, Lindzey JK, Prescott JL, Kumar M V, Tindall DJ. The androgen receptor in the testicular feminized (Tfm) mouse may be a product of internal translation initiation. Receptor 1994;4(2):121–134. http://www.ncbi.nlm.nih.gov/pubmed/7950980 [PubMed] [Google Scholar]
- 135. Politch JA, Fox TO, Houben P, Bullock L, Lovell D. TfmLac: a second isolation of testicular feminization in mice. Biochem Genet. 1988;26(3–4):213‐221. doi: 10.1007/BF00561461 [DOI] [PubMed] [Google Scholar]
- 136. Kerkhofs S, Denayer S, Haelens A, Claessens F. Androgen receptor knockout and knock‐in mouse models. J Mol Endocrinol. 2009;42(1):11‐17. doi: 10.1677/JME-08-0122 [DOI] [PubMed] [Google Scholar]
- 137. Matsumoto T, Takeyama KI, Sato T, Kato S. Androgen receptor functions from reverse genetic models. J Steroid Biochem Mol Biol. 2003;85(2–5):95‐99. doi: 10.1016/S0960-0760(03)00231-0 [DOI] [PubMed] [Google Scholar]
- 138. Fan WQ, Yanase T, Nomura M, et al. Androgen receptor null male mice develop late‐onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion. Diabetes. 2005;54(4):1000‐1008. doi: 10.2337/diabetes.54.4.1000 [DOI] [PubMed] [Google Scholar]
- 139. Sato T, Matsumoto T, Yamada T, Watanabe T, Kawano H, Kato S. Late onset of obesity in male androgen receptor‐deficient (AR KO) mice. Biochem Biophys Res Commun. 2003;300(1):167‐171. doi: 10.1016/S0006-291X(02)02774-2 [DOI] [PubMed] [Google Scholar]
- 140. Yeh S, Tsai MY, Xu Q, et al. Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci U S A. 2002;99(21):13498‐13503. doi: 10.1073/pnas.212474399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Yu IC, Lin HY, Liu NC, et al. Hyperleptinemia without obesity in male mice lacking androgen receptor in adipose tissue. Endocrinology. 2008;149(5):2361‐2368. doi: 10.1210/en.2007-0516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Lin H‐Y, Xu Q, Yeh S, Wang R‐S, Sparks JD, Chang C. Insulin and leptin resistance with hyperleptinemia in mice lacking androgen receptor. Diabetes. 2005;54(6):1717‐1725. doi: 10.2337/diabetes.54.6.1717 [DOI] [PubMed] [Google Scholar]
- 143. De Gendt K, Swinnen JV, Saunders PTK, et al. A Sertoli cell‐selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci U S A. 2004;101(5):1327‐1332. doi: 10.1073/pnas.0308114100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Rana K, Fam BC, Clarke MV, Pang TPSS, Zajac JD, Maclean HE. Increased adiposity in DNA binding‐dependent androgen receptor knockout male mice associated with decreased voluntary activity and not insulin resistance. Am J Physiol ‐ Endocrinol Metab. 2011;301(5):767‐778. doi: 10.1152/ajpendo.00584.2010 [DOI] [PubMed] [Google Scholar]
- 145. Notini AJ, Davey RA, McManus JF, Bate KL, Zajac JD. Genomic actions of the androgen receptor are required for normal male sexual differentiation in a mouse model. J Mol Endocrinol. 2005;35(3):547‐555. doi: 10.1677/jme.1.01884 [DOI] [PubMed] [Google Scholar]
- 146. Wu J, Henning P, Sjögren K, et al. The androgen receptor is required for maintenance of bone mass in adult male mice. Mol Cell Endocrinol. 2019;479(May 2018):159‐169. doi: 10.1016/j.mce.2018.10.008 [DOI] [PubMed] [Google Scholar]
- 147. Sato T, Matsumoto T, Kawano H, et al. Brain masculinization requires androgen receptor function. Proc Natl Acad Sci U S A. 2004;101(6):1673‐1678. doi: 10.1073/pnas.0305303101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. De Gendt K, Verhoeven G. Tissue‐ and cell‐specific functions of the androgen receptor revealed through conditional knockout models in mice. Mol Cell Endocrinol. 2012;352(1–2):13‐25. doi: 10.1016/j.mce.2011.08.008 [DOI] [PubMed] [Google Scholar]
- 149. Goldstein SR, Siddhanti S, Ciaccia AV, Plouffe L. A pharmacological review of selective oestrogen receptor modulators. Hum Reprod Update. 2000;6(3):212‐224. doi: 10.1093/humupd/6.3.212 [DOI] [PubMed] [Google Scholar]
- 150. McInnes KJ, Smith LB, Hunger NI, Saunders PTK, Andrew R, Walker BR. Deletion of the androgen receptor in adipose tissue in male mice elevates retinol binding protein 4 and reveals independent effects on visceral fat mass and on glucose homeostasis. Diabetes. 2012;61(5):1072‐1081. doi: 10.2337/db11-1136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Clarke MV, Russell PK, Zajac JD, Davey RA. The androgen receptor in the hypothalamus positively regulates hind‐limb muscle mass and voluntary physical activity in adult male mice. J Steroid Biochem Mol Biol. 2019;189(December 2018):187‐194. doi: 10.1016/j.jsbmb.2019.02.018 [DOI] [PubMed] [Google Scholar]
- 152. Ophoff J, Van Proeyen K, Callewaert F, et al. Androgen signaling in myocytes contributes to the maintenance of muscle mass and fiber type regulation but not to muscle strength or fatigue. Endocrinology. 2009;150(8):3558‐3566. doi: 10.1210/en.2008-1509 [DOI] [PubMed] [Google Scholar]
- 153. Dubois V, Laurent MR, Sinnesael M, et al. A satellite cell‐specific knockout of the androgen receptor reveals myostatin as a direct androgen target in skeletal muscle. FASEB J. 2014;28(7):2979‐2994. doi: 10.1096/fj.14-249748 [DOI] [PubMed] [Google Scholar]
- 154. Cipriani C, Colangelo L, Santori R, et al. The interplay between bone and glucose metabolism. Front Endocrinol (Lausanne). 2020;11(March):1‐9. doi: 10.3389/fendo.2020.00122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Zhou R, Guo Q, Xiao Y, et al. Endocrine role of bone in the regulation of energy metabolism. Bone Res. 2021;9:25. doi: 10.1038/s41413-021-00142-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Karsenty G, Kousteni S. Bone as an endocrine organ. Encycl Endocr Dis. 2018;18(5):47‐51. doi: 10.1016/B978-0-12-801238-3.64343-X [DOI] [Google Scholar]
- 157. Rubinow KB, Wang S, Den Hartigh LJ, et al. Hematopoietic androgen receptor deficiency promotes visceral fat deposition in male mice without impairing glucose homeostasis. Andrology. 2015;3(4):787‐796. doi: 10.1111/andr.12055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Wiren KM, Hashimoto JG, Zhang XW. Stem cell activation in adults can reverse detrimental changes in body composition to reduce fat and increase lean mass in both sexes. J Cell Biochem. 2011;112(12):3638‐3647. doi: 10.1002/jcb.23288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Wiren KM, Zhang XW, Toombs AR, et al. Targeted overexpression of androgen receptor in osteoblasts: unexpected complex bone phenotype in growing animals. Endocrinology. 2004;145(7):3507‐3522. doi: 10.1210/en.2003-1016 [DOI] [PubMed] [Google Scholar]
- 160. Rubinow K, Houston B, Wang S, et al. Androgen receptor deficiency in monocytes/macrophages does not alter adiposity or glucose homeostasis in male mice. Asian J Androl. 2018;20(3):276‐283. doi: 10.4103/aja.aja_54_17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Bianco P, Kuznetsov SA, Riminucci M, Gehron RP. Postnatal Skeletal Stem Cells. Methods Enzymol. 2006;419(06):117‐148. doi: 10.1016/S0076-6879(06)19006-0 [DOI] [PubMed] [Google Scholar]
- 162. Huang CK, Lai KP, Luo J, et al. Loss of androgen receptor promotes adipogenesis but suppresses osteogenesis in bone marrow stromal cells. Stem Cell Res. 2013;11(2):938‐950. doi: 10.1016/j.scr.2013.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Robling AG, Castillo AB, Turner CH. Biomechanical and molecular regulation of bone remodeling. Annu Rev Biomed Eng. 2006;8(May):455‐498. doi: 10.1146/annurev.bioeng.8.061505.095721 [DOI] [PubMed] [Google Scholar]
- 164. Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Horm Mol Biol Clin Investig. 2016;28(1):21‐38. doi: 10.1515/hmbci-2016-0012 [DOI] [PubMed] [Google Scholar]
- 165. Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone—the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res. 2018;6(1):1‐11. doi: 10.1038/s41413-018-0019-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Semirale AA, Zhang XW, Wiren KM. Body composition changes and inhibition of fat development in vivo implicates androgen in regulation of stem cell lineage allocation. J Cell Biochem. 2011;112(7):1773‐1786. doi: 10.1002/jcb.23098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Atlantis E, Martin SA, Haren MT, et al. Demographic, physical and lifestyle factors associated with androgen status: the Florey Adelaide Male Ageing Study (FAMAS). Clin Endocrinol (Oxf). 2009;71(2):261‐272. doi: 10.1111/j.1365-2265.2008.03463.x [DOI] [PubMed] [Google Scholar]
- 168. Wilson JD, Griffin JE, George FW, Leshin M. The endocrine control of male phenotypic development. Aust J Biol Sci. 1983;36(2):101‐128. doi: 10.1071/BI9830101 [DOI] [PubMed] [Google Scholar]
- 169. Song Y, Tangen C, Goodman P, et al. Finasteride, prostate cancer, and weight gain: Evidence for genetic or environmental factors that affect cancer outcomes during finasteride treatment. Prostate. 2008;68:281‐286. 10.1002/pros.20637 [DOI] [PubMed] [Google Scholar]
- 170. Bhasin S, Travison TG, Storer TW, et al. Effect of testosterone supplementation with and without a dual 5α‐reductase inhibitor on fat‐free mass in men with suppressed testosterone production: a randomized controlled trial. JAMA ‐ J Am Med Assoc. 2012;307(9):931‐939. doi: 10.1001/jama.2012.227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Page ST, Amory JK, Bowman FDB, et al. Exogenous testosterone (T) alone or with finasteride increases physical performance, grip strength, and lean body mass in older men with low serum T. J Clin Endocrinol Metab. 2005;90(3):1502‐1510. doi: 10.1210/jc.2004-1933 [DOI] [PubMed] [Google Scholar]
- 172. 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. J Sex Med. 2014;11(2):563‐573. doi: 10.1111/jsm.12368 [DOI] [PubMed] [Google Scholar]
- 173. Gruntmanis U. The role of 5α‐reductase inhibition in men receiving testosterone replacement therapy. JAMA ‐ J am Med Assoc. 2012;307(9):968‐970. doi: 10.1001/jama.2012.259 [DOI] [PubMed] [Google Scholar]
- 174. Idan A, Griffiths KA, Harwood DT, et al. Long‐term effects of dihydrotestosterone treatment on prostate growth in healthy, middle‐aged men without prostate disease: a randomized, placebo‐controlled trial. Ann Intern Med. 2010;153(10):621‐632. doi: 10.7326/0003-4819-153-10-201011160-00004 [DOI] [PubMed] [Google Scholar]
- 175. Kim NR, David K, Corbeels K, et al. Testosterone reduces body fat in male mice by stimulation of physical activity via extrahypothalamic ERα signaling. Endocrinology. 2021;0(0):1‐14. doi: 10.1210/endocr/bqab045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Livingstone DEW, Barat P, Di Rollo EM, et al. 5α‐Reductase type 1 deficiency or inhibition predisposes to insulin resistance, hepatic steatosis, and liver fibrosis in rodents. Diabetes. 2015;64(2):447‐458. doi: 10.2337/db14-0249 [DOI] [PubMed] [Google Scholar]
- 177. Windahl SH, Andersson N, Börjesson AE, et al. Reduced bone mass and muscle strength in male 5α‐reductase type 1 inactivated mice. PLoS ONE. 2011;6(6):1‐8. doi: 10.1371/journal.pone.0021402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Movérare‐Skrtic S, Venken K, Andersson N, et al. Dihydrotestosterone treatment results in obesity and altered lipid metabolism in orchidectomized mice. Obesity. 2006;14(4):662‐672. doi: 10.1038/oby.2006.75 [DOI] [PubMed] [Google Scholar]
- 179. Rubinow KB. Estrogens and body weight regulation in men. Adv Exp Med Biol. 2017;1043:285‐313. doi: 10.1007/978-3-319-70178-3_14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Jones MEE, Chin Boon W, Proietto J, Simpson ER. Of mice and men: the evolving phenotype of aromatase deficiency. Trends Endocrinol Metab. 2006;17(2):55‐64. doi: 10.1016/j.tem.2006.01.004 [DOI] [PubMed] [Google Scholar]
- 181. Finkelstein JS, Lee H, Burnett‐Bowie S‐AM, et al. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med. 2013;369(11):1011‐1022. doi: 10.1056/nejmoa1206168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. de Ronde R, de Jong FH. Aromatase inhibitors in men: effects and therapeutic options. Reprod Biol Endocrinol. 2011;9:93. doi: 10.1186/1477-7827-9-93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Awouters M, Vanderschueren D, Antonio L. Aromatase inhibitors and selective estrogen receptor modulators: unconventional therapies for functional hypogonadism? Andrology. 2020;8(6):1590‐1597. doi: 10.1111/andr.12725 [DOI] [PubMed] [Google Scholar]
- 184. Amano A, Kondo Y, Noda Y, et al. Abnormal lipid/lipoprotein metabolism and high plasma testosterone levels in male but not female aromatase‐knockout mice. Arch Biochem Biophys. 2017;622:47‐58. doi: 10.1016/j.abb.2017.03.007 [DOI] [PubMed] [Google Scholar]
- 185. Jones MEE, Thorburn AW, Britt KL, et al. Aromatase‐deficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci. 2000;97(23):12735‐12740. doi: 10.1073/pnas.97.23.12735 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Fisher CR, Graves KH, Parlow AF, Simpson ER. Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proc Natl Acad Sci U S A. 1998;95(12):6965‐6970. doi: 10.1073/pnas.95.12.6965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Takeda K, Toda K, Saibara T, et al. Progressive development of insulin resistance phenotype in male mice with complete aromatase (CYP19) deficiency. J Endocrinol. 2003;176(2):237‐246. doi: 10.1677/joe.0.1760237 [DOI] [PubMed] [Google Scholar]
- 188. Van Sinderen ML, Steinberg GR, Jørgensen SB, et al. Hepatic glucose intolerance precedes hepatic steatosis in the male aromatase knockout (ArKO) mouse. PLoS ONE. 2014;9(2):1‐11. doi: 10.1371/journal.pone.0087230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Rubinow KB, den Hartigh LJ, Goodspeed L, Wang S, Oz OK. Aromatase deficiency in hematopoietic cells improves glucose tolerance in male mice through skeletal muscle‐specific effects. PLoS ONE. 2020;15(1):1‐18. doi: 10.1371/journal.pone.0227830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Ohlsson C, Hammarstedt A, Vandenput L, et al. Increased adipose tissue aromatase activity improves insulin sensitivity and reduces adipose tissue inflammation in male mice. Am J Physiol ‐ Endocrinol Metab. 2017;313(4):E450‐E462. doi: 10.1152/ajpendo.00093.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Stocco C. Tissue physiology and pathology of aromatase. Steroids. 2012;77(1–2):27‐35. doi: 10.1016/j.steroids.2011.10.013.Tissue [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Taylor AH, Al‐Azzawi F. Immunolocalisation of oestrogen receptor beta in human tissues. J Mol Endocrinol. 2000;24(1):145‐155. doi: 10.1677/jme.0.0240145 [DOI] [PubMed] [Google Scholar]
- 193. Prossnitz ER, Barton M. The G‐protein‐coupled estrogen receptor GPER in health and disease. Nat Rev Endocrinol. 2011;7(12):715‐726. doi: 10.1038/nrendo.2011.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Frank AP, De Souza SR, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity‐related health risks. J Lipid Res. 2019;60(10):1710‐1719. doi: 10.1194/jlr.R086975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Blüher M. Importance of estrogen receptors in adipose tissue function. Mol Metab. 2013;2(3):130‐132. doi: 10.1016/j.molmet.2013.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Smith EP, Boyd J, Frank GR, et al. Estrogen resistance caused by a mutation in the estrogen‐receptor gene in a man. N Engl J Med. 1994;331(16):1056‐1061. doi: 10.1056/NEJM199410203311604 [DOI] [PubMed] [Google Scholar]
- 197. Bernard V, Kherra S, Francou B, et al. Familial multiplicity of estrogen insensitivity associated with a loss‐of‐function ESR1 mutation. J Clin Endocrinol Metab. 2017;102(1):93‐99. doi: 10.1210/jc.2016-2749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Colleluori G, Chen R, Turin CG, et al. Aromatase inhibitors plus weight loss improves the hormonal profile of obese hypogonadal men without causing major side effects. Front Endocrinol (Lausanne). 2020;11(May):1‐12. doi: 10.3389/fendo.2020.00277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Dias JP, Melvin D, Simonsick EM, et al. Effects of aromatase inhibition vs. testosterone in older men with low testosterone: randomized‐controlled trial. Andrology. 2016;4(1):33‐40. doi: 10.1111/andr.12126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Rambhatla A, Mills JN, Rajfer J. The role of estrogen modulators in male hypogonadism and infertility. Rev Urol. 2016;18(2):66‐72. doi: 10.3909/riu0711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. V. Bendre S, J. Murray P. Clomiphene citrate effectively increases testosterone in obese, young, hypogonadal men. Reprod Syst Sex Disord. 2015;04(04):1‐8. doi: 10.4172/2161-038x.1000155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Pelusi C, Giagulli VA, Baccini M, et al. Clomiphene citrate effect in obese men with low serum testosterone treated with metformin due to dysmetabolic disorders: a randomized, double‐blind, placebo‐controlled study. PLoS ONE. 2017;12(9):1‐16. doi: 10.1371/journal.pone.0183369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Soares AH, Horie NC, Chiang LAP, et al. Effects of clomiphene citrate on male obesity‐associated hypogonadism: a randomized, double‐blind, placebo‐controlled study. Int J Obes (Lond). 2018;42(5):953‐963. doi: 10.1038/s41366-018-0105-2 [DOI] [PubMed] [Google Scholar]
- 204. Park CJ, Zhao Z, Glidewell‐Kenney C, et al. Genetic rescue of nonclassical ERα signaling normalizes energy balance in obese Erα‐null mutant mice. J Clin Invest. 2011;121(2):604‐612. doi: 10.1172/JCI41702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor‐α knockout mice. Proc Natl Acad Sci USA. 2000;97(23):12729‐12734. doi: 10.1073/pnas.97.23.12729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Couse JF, Korach KS. Erratum: Estrogen receptor null mice: what have we learned and where will they lead us? Endocr Rev. 1999;20(3):358–417. [DOI] [PubMed] [Google Scholar]
- 207. Ohlsson C, Hellberg N, Parini P, et al. Obesity and disturbed lipoprotein profile in estrogen receptor‐α‐deficient male mice. Biochem Biophys Res Commun. 2000;278(3):640‐645. doi: 10.1006/bbrc.2000.3827 [DOI] [PubMed] [Google Scholar]
- 208. Iñigo MR, Amorese AJ, Tarpey MD, et al. Estrogen receptor‐α in female skeletal muscle is not required for regulation of muscle insulin sensitivity and mitochondrial regulation. Mol Metab. 2020;34(December 2019):1‐15. doi: 10.1016/j.molmet.2019.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Bryzgalova G, Gao H, Ahren B, et al. Evidence that oestrogen receptor‐α plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver. Diabetologia. 2006;49(3):588‐597. doi: 10.1007/s00125-005-0105-3 [DOI] [PubMed] [Google Scholar]
- 210. Davis KE, D. Neinast M, Sun K, et al. 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. doi: 10.1016/j.molmet.2013.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Windahl SH, Börjesson AE, Farman HH, et al. Estrogen receptor‐α in osteocytes is important for trabecular bone formation in male mice. Proc Natl Acad Sci U S A. 2013;110(6):2294‐2299. doi: 10.1073/pnas.1220811110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Zhu L, Shi J, Luu TN, et al. Hepatocyte estrogen receptor alpha mediates estrogen action to promote reverse cholesterol transport during Western‐type diet feeding. Mol Metab. 2018;8(December 2017):106‐116. doi: 10.1016/j.molmet.2017.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Yepuru M, Eswaraka J, Kearbey JD, et al. Estrogen receptor‐β‐selective ligands alleviate high‐fat diet‐ and ovariectomy‐induced obesity in mice. J Biol Chem. 2010;285(41):31292‐31303. doi: 10.1074/jbc.M110.147850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. González‐Granillo M, Savva C, Li X, et al. Selective estrogen receptor (ER)β activation provokes a redistribution of fat mass and modifies hepatic triglyceride composition in obese male mice. Mol Cell Endocrinol. 2020;502(December 2019):110672. doi: 10.1016/j.mce.2019.110672 [DOI] [PubMed] [Google Scholar]
- 215. Luo F, Huang WY, Guo Y, Ruan GY, Peng R, Li XP. 17Β‐estradiol lowers triglycerides in adipocytes via estrogen receptor α and it may be attenuated by inflammation. Lipids Health Dis. 2017;16(1):1‐6. doi: 10.1186/s12944-017-0575-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Yasrebi A, Rivera JA, Krumm EA, Yang JA, Roepke TA. Activation of estrogen response element‐independent ERα signaling protects female mice from diet‐induced obesity. Endocrinology. 2017;158(2):319‐334. doi: 10.1210/en.2016-1535 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Hautanen A. Synthesis and regulation of sex hormone‐binding globulin in obesity. Int J Obes (Lond). 2000;24:S64‐S70. doi: 10.1038/sj.ijo.0801281 [DOI] [PubMed] [Google Scholar]
- 218. Haffner S. Sex hormones, obesity, fat distribution, type 2 diabetes and insulin resistance: epidemiological and clinical correlation. Int J Obes. 2000;24:S56‐S58. doi: 10.1038/sj/ijo/0801279 [DOI] [PubMed] [Google Scholar]
- 219. Hammond GL, Hogeveen KN. Sex hormone‐binding globulin (SHBG). In: Henry HL, Norman AW, eds. Encyclopedia of Hormones. New York: Academic Press; 2003:340‐344. 10.1016/B0-12-341103-3/00269-2 [DOI] [Google Scholar]
- 220. Hansson V, Weddington SC, French FS, et al. Secretion and role of androgen‐binding proteins in the testis and epididymis. J Reprod Fertil Suppl. 1976;(24 suppl):17–33. https://pubmed.ncbi.nlm.nih.gov/1069850/ [PubMed] [Google Scholar]
- 221. Joseph DR, Sullivan PM, Wang YM, Millhorn DE, Bayliss DM. Complex structure and regulation of the ABP/SHBG gene. J Steroid Biochem Mol Biol. 1991;40(4–6):771‐775. doi: 10.1016/0960-0760(91)90302-L [DOI] [PubMed] [Google Scholar]
- 222. Gyawali P, Martin SA, Heilbronn LK, et al. Cross‐sectional and longitudinal determinants of serum sex hormone binding globulin (SHBG) in a cohort of community‐dwelling men. PLoS ONE. 2018;13(7):1‐15. doi: 10.1371/journal.pone.0200078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Roberts CK, Croymans DM, Aziz N, Butch AW, Lee CC. Resistance training increases SHBG in overweight/obese, young men. Metabolism. 2013;62(5):725‐733. doi: 10.1016/j.metabol.2012.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Boonchaya‐anant P, Laichuthai N, Suwannasrisuk P, Houngngam N, Udomsawaengsup S, Snabboon T. Changes in testosterone levels and sex hormone‐binding globulin levels in extremely obese men after bariatric surgery. Int J Endocrinol. 2016;2016:1416503. doi: 10.1155/2016/1416503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Kim C, Dabelea D, Kalyani RR, et al. Changes in visceral adiposity, subcutaneous adiposity, and sex hormones in the diabetes prevention program. J Clin Endocrinol Metab. 2017;102(9):3381‐3389. doi: 10.1210/jc.2017-00967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Laurent MR, Hammond GL, Blokland M, et al. Sex hormone‐binding globulin regulation of androgen bioactivity in vivo: validation of the free hormone hypothesis. Sci Rep. 2016;6(May):1‐12. doi: 10.1038/srep35539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Jänne M, Deol HK, Power SGA, Yee SP, Hammond GL. Human sex hormone‐binding globulin gene expression in transgenic mice. Mol Endocrinol. 1998;12(1):123‐136. doi: 10.1210/mend.12.1.0050 [DOI] [PubMed] [Google Scholar]
- 228. Saéz‐López C, Rivera‐Giménez M, Hernández C, Simó R, Selva DM. SHBG‐C57BL/ksJ‐db/db: a new mouse model to study SHBG expression and regulation during obesity development. Endocrinology. 2015;156(12):4571‐4581. doi: 10.1210/en.2015-1677 [DOI] [PubMed] [Google Scholar]
- 229. Saez‐Lopez C, Villena JA, Simó R, Selva DM. Sex hormone‐binding globulin overexpression protects against high‐fat diet‐induced obesity in transgenic male mice. J Nutr Biochem. 2020;85:108480. doi: 10.1016/j.jnutbio.2020.108480 [DOI] [PubMed] [Google Scholar]
- 230. Sofer Y, Nevo N, Vechoropoulos M, et al. Human sex hormone‐binding globulin does not provide metabolic protection against diet‐induced obesity and dysglycemia in mice. Endocr Connect. 2018;7(1):91‐96. doi: 10.1530/EC-17-0240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Srivastava G, Apovian C. Future pharmacotherapy for obesity: new anti‐obesity drugs on the horizon. Curr Obes Rep. 2018;7(2):147‐161. doi: 10.1007/s13679-018-0300-4 [DOI] [PubMed] [Google Scholar]
- 232. Dimri M, Satyanarayana A. Molecular signaling pathways and therapeutic targets in hepatocellular carcinoma. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Schweizer L, Zhang L. Enhancing cancer drug discovery through novel cell signaling pathway panel strategy.:53–59. 10.4137/CGM.S11134 [DOI] [PMC free article] [PubMed]
- 234. Zhang L, Decicco C. The changing face of innovation in drug discovery. Collab Innov Drug Discov Strateg Public Priv Partnerships. 2014;31‐56. doi: 10.1002/9781118778166.ch4 [DOI] [Google Scholar]
- 235. Rana K, Chiu MWS, Russell PK, et al. Muscle‐specific androgen receptor deletion shows limited actions in myoblasts but not in myofibers in different muscles in vivo. J Mol Endocrinol. 2016;57(2):125‐138. doi: 10.1530/JME-15-0320 [DOI] [PubMed] [Google Scholar]
- 236. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol. 2012;8(8):457‐465. doi: 10.1038/nrendo.2012.49 [DOI] [PubMed] [Google Scholar]
- 237. Zorena K, Jachimowicz‐Duda O, Ślęzak D, Robakowska M, Mrugacz M. Adipokines and obesity. Potential link to metabolic disorders and chronic complications. Int J Mol Sci. 2020;21(10):3570. doi: 10.3390/ijms21103570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Cheleuitte D, Mizuno S, Glowacki J. In vitro secretion of cytokines by human bone marrow: effects of age and estrogen status. J Clin Endocrinol Metab. 1998;83(6):2043‐2051. doi: 10.1210/jc.83.6.2043 [DOI] [PubMed] [Google Scholar]
- 239. Tencerova M, Kassem M. The bone marrow‐derived stromal cells: commitment and regulation of adipogenesis. Front Endocrinol. 2016;7. doi: 10.3389/fendo.2016.00127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Nishizawa H, Shimomura L, Kishida K, et al. Androgens decrease plasma adiponectin, an insulin‐sensitizing adipocyte‐derived protein. Diabetes. 2002;51(9):2734‐2741. doi: 10.2337/diabetes.51.9.2734 [DOI] [PubMed] [Google Scholar]
- 241. Jockenhövel F, Blum WF, Vogel E, et al. Testosterone substitution normalizes elevated serum leptin levels in hypogonadal men. J Clin Endocrinol Metab. 1997;82(8):2510‐2513. doi: 10.1210/jcem.82.8.4174 [DOI] [PubMed] [Google Scholar]
- 242. Castillo‐Armengol J, Fajas L, Lopez‐Mejia IC. Inter‐organ communication: a gatekeeper for metabolic health. EMBO Rep. 2019;20(9):1‐16. doi: 10.15252/embr.201947903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Guo A, Li K, Xiao Q. Sarcopenic obesity: myokines as potential diagnostic biomarkers and therapeutic targets? Exp Gerontol. 2020;139(June):111022. doi: 10.1016/j.exger.2020.111022 [DOI] [PubMed] [Google Scholar]
- 244. Blüher M. Adipokines—removing road blocks to obesity and diabetes therapy. Mol Metab. 2014;3(3):230‐240. doi: 10.1016/j.molmet.2014.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Montgomery MK, Bayliss J, Devereux C, et al. SMOC1 is a glucose‐responsive hepatokine and therapeutic target for glycemic control. Sci Transl Med. 2020;12(559):1‐14. doi: 10.1126/SCITRANSLMED.AAZ8048 [DOI] [PubMed] [Google Scholar]
- 246. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(DECEMBER):425‐432. [DOI] [PubMed] [Google Scholar]
- 247. Peinetti N, Cuello Rubio MM, Sosa LDV, et al. Testosterone‐loaded GM1 micelles targeted to the intracellular androgen receptor for the specific induction of genomic androgen signaling. Int J Pharm. 2020;591:119985. doi: 10.1016/j.ijpharm.2020.119985 [DOI] [PubMed] [Google Scholar]
- 248. Solomon ZJ, Mirabal JR, Mazur DJ, Kohn TP, Lipshultz LI, Pastuszak AW. Selective androgen receptor modulators: current knowledge and clinical applications. Sex Med Rev. 2019;7(1):84‐94. doi: 10.1016/j.sxmr.2018.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Narayanan R, Coss CC, Dalton JT. Development of selective androgen receptor modulators (SARMs). Mol Cell Endocrinol. 2018;465(1):134‐142. doi: 10.1016/j.mce.2017.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Hruby A, Hu FB. The epidemiology of obesity: a big picture. Pharmacoeconomics. 2015;33(7):673‐689. doi: 10.1007/s40273-014-0243-x [DOI] [PMC free article] [PubMed] [Google Scholar]
