Next Article in Journal
Immunomodulation of Mesenchymal Stem Cells in Acute Lung Injury: From Preclinical Animal Models to Treatment of Severe COVID-19
Next Article in Special Issue
Targeting ABCC6 in Mesenchymal Stem Cells: Impairment of Mature Adipocyte Lipid Homeostasis
Previous Article in Journal
Reorganization of the Brain Extracellular Matrix in Hippocampal Sclerosis
Previous Article in Special Issue
Adipose Tissue Inflammation and Pulmonary Dysfunction in Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adipose Tissue Dysfunction and Obesity-Related Male Hypogonadism

Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(15), 8194; https://doi.org/10.3390/ijms23158194
Submission received: 10 June 2022 / Revised: 19 July 2022 / Accepted: 21 July 2022 / Published: 25 July 2022

Abstract

:
Obesity is a chronic illness associated with several metabolic derangements and comorbidities (i.e., insulin resistance, leptin resistance, diabetes, etc.) and often leads to impaired testicular function and male subfertility. Several mechanisms may indeed negatively affect the hypothalamic–pituitary–gonadal health, such as higher testosterone conversion to estradiol by aromatase activity in the adipose tissue, increased ROS production, and the release of several endocrine molecules affecting the hypothalamus–pituitary–testis axis by both direct and indirect mechanisms. In addition, androgen deficiency could further accelerate adipose tissue expansion and therefore exacerbate obesity, which in turn enhances hypogonadism, thus inducing a vicious cycle. Based on these considerations, we propose an overview on the relationship of adipose tissue dysfunction and male hypogonadism, highlighting the main biological pathways involved and the current therapeutic options to counteract this condition.

1. Introduction

Obesity, a worldwide health problem, is characterized by excessive fat accumulation. Globally, the prevalence of obesity has nearly tripled since 1975, with about 13% of adults being obese, and it will continue to rise rapidly in the next years. Indeed, one in five adults are predicted to have obesity by 2025 [1]. Obesity is also one of the key risk factors for many noncommunicable diseases, such as coronary heart disease, hypertension and stroke, certain types of cancer, type 2 diabetes, gallbladder disease, dyslipidemia, osteoarthritis and gout, and pulmonary diseases, including sleep apnea, and represents one of the principal causes of secondary hypogonadism in men, a condition characterized by the impairment of the hypothalamic–pituitary–testicular (HPT) axis, in which the reduction in testosterone levels is also accompanied by signs and symptoms of hypogonadism, such as decreases in libido, erectile function, semen quality, strength, and mood [2,3,4,5,6]. The prevalence of hypogonadism in normal-weight adult males is around 32%, while about 75% of subjects with severe obesity (BMI > 40 kg/m2) have from hypogonadism [7,8,9,10].
Under physiological conditions, the HPT axis is activated by kisspeptins, peptides able to regulate the secretion of hypothalamic gonadotropin-releasing hormone (GnRH) into the hypothalamic–hypophyseal portal circulation, thus stimulating the release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the pituitary gland. Both hormones travel in the systemic circulation and reach the testes, where LH stimulates the secretion of testosterone from Leydig cells and, along with FSH, supports appropriate spermatogenesis [11]. Obese subjects exhibit decreased levels of total and free testosterone as compared to lean subjects [12], and both BMI and waist circumference are strongly related to the degree of HPT axis dysfunction [13,14]. Furthermore, hypogonadism is associated with fat accumulation, leading to an endless loop in which abnormal adipose tissue (AT) expansion impairs testosterone production, resulting in further accumulation of AT [11].
Several mechanisms may favor hypogonadism; these are mainly related to the excessive amount of AT, particularly in the visceral depot, and to the derangement of its function [15,16]. The impact and contribution of these on the development of obesity-related hypogonadism are not fully elucidated. Herein, we summarize emerging in vivo and in vitro evidence supporting the link between obesity and hypogonadism and the effects of both pharmacological and non-pharmacological strategies to contrast obesity and restore the eugonadal state.

2. The Effects of Adipose Tissue Dysfunction on Androgen Levels

2.1. Direct Effects of Adipose Tissue

Obesity is a consequence of the increased accumulation of lipids and expansion of AT. As testosterone is a fat-soluble molecule, we can speculate that it is likely to be sequestered into AT depots, leading to reduced circulating levels of this substance in obesity; however, to date, the measurement of steroid concentrations in fat cells and tissue has produced conflicting results [17,18,19]. Indeed, adipocytes from subcutaneous AT (SAT) retrieved from obese men were shown to have higher concentrations of intracellular testosterone as compared with those retrieved from lean men [20]. Despite this greater testosterone accumulation, SAT from obese patients had lower expression of androgen-responsive genes involved in the lipolytic and anti-adipogenic pathways, suggesting the altered function of adipose cells in these individuals. This may, in turn, lead to reduced circulating testosterone levels and reduced fat oxidation in obese men [20].
AT from obese subjects also shows an upregulation of aromatase activity, which is proportional to body fat mass and converts testosterone into estrogens. The increased levels of estrogens, in turn, reduce LH pulse amplitude and may directly upregulate adipogenesis and increase subcutaneous, ectopic, and visceral fat [21,22]. Therefore, the obesity-induced increase in aromatase expression may lead to further peripheral fat accumulation, both by increasing the concentration of estrogens and reducing LH-induced testosterone production [22,23].
Taken together, these results suggest that AT may participate directly in androgen deprivation both by sequestering testosterone from the systemic circulation and through its conversion to estradiol, and indirectly by reducing the hypothalamic stimulus on testosterone synthesis.

2.2. Effects of Adipokines

Obesity is also known to alter the release of adipocytokines from AT, resulting in important health consequences [24,25,26]. For instance, the increased secretion of leptin by expanded AT is known to impair central leptin signaling and to participate in the development of metabolic alterations and HPT dysfunction [27,28,29]. Indeed, hyperleptinemia, typical in obesity, is associated with the onset of androgen deficiency and abnormalities of the reproductive tract, sperm count, and motility [15,30,31].
Leptin regulates HPT functionality both through indirect control of forebrain neurons and direct control of testicular tissue. Early experimental studies revealed that kisspeptin neurons are targets of leptin activity. Indeed, leptin-deficient obese male mice show a reduction in kisspeptin production. The levels of kisspeptin were restored, even if only partially, when mice were treated with leptin replacement therapy, and this was associated with an increase in the cellular content of KISS-1 mRNA by 28% [32]. Specifically, both leptin and kisspeptin are co-expressed in neurons of the arcuate nucleus (ARC) of the hypothalamus, the action of which appears to mediate a negative effect on sex steroids and gonadotropin secretion via the control of GnRH pulses at or near GnRH nerve terminals [32,33,34,35] (Figure 1).
Beyond the central regulation of the GnRH pathways, leptin appears to regulate male reproduction by regulating testicular cells. Indeed, leptin can cross the blood–testis barrier, thus modulating the steroidogenic process [36]. In males, the expression of the leptin receptor (LEPR) in the gonads appears to be restricted to Leydig cells; its levels are inversely correlated with testosterone concentration and with abnormalities of sperm quality and production [37]. Leptin acts on testicular tissue principally through the activation of the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway [38,39]. Briefly, the activation of JAK2 causes the phosphorylation of several residues of LEPR, which consequently leads to the activation of STAT3, resulting in nuclear translocation and steroid gene transcription (i.e., translocator protein [TSPO], steroidogenic acute regulatory protein [STAR]), or the recruitment of suppressor of cytokine signaling 3 (SOCS3), the key inhibitor of leptin signaling [40,41]. In obesity and hyperleptinemia, negative feedback control of leptin signaling occurs due to the increased expression of SOCS3 inhibiting the phosphorylation of JAK2, thus contributing to the onset of leptin resistance [42] (Figure 1). In this setting, a variation in reproductive parameters occurs, as reported in hyperleptinemic mice, in which reduced in testosterone levels, in association with a decrease in the volume and weight of testes, as well as in the number of spermatocytes, sperm, Leydig cells, and offspring, were noted [43]. All these effects were principally mediated by the upregulation of SOCS3 resulting in the inhibition of JAK2 phosphorylation, showing that this pathway has a pivotal role in the control of androgen levels and reproductive performance [43]. Moreover, Zaho et al. confirmed that both short and prolonged exposure to a high-fat diet (HFD) increases circulating leptin levels in mice together with apoptosis in Leydig cells and reduced sperm count and motility, which are all events correlated with increased oxidative stress in the testes [43,44].
In vitro data also established that supraphysiological levels of leptin inhibited steroidogenic gene expression and the activation of the JAK/STAT pathway in a tumoral line of murine Leydig cells [45]. The same pathway was found to be involved in central regulation of leptin signaling as displayed in mice with neural-specific disruption of STAT3, which recapitulated the phenotype typically observed in leptin resistance (i.e., hyperphagia, diabetes) in association with an increase in infertility [46]. Together, these results indicate that adequate leptin signaling in both testes and neuronal circuits sustains the physiological activity of the HPT axis, and that the presence of leptin-resistance could favor the development of functional hypogonadism and the related reproductive dysfunction.
Among the humoral factors secreted by AT, adiponectin was recently found to regulate the expression of genes implicated in steroidogenesis. Adiponectin release was found to be reduced in the presence of obesity, insulin resistance, and other co-morbidities [47,48,49]. More recently, the downregulation of adiponectin was observed in association with reduced testosterone levels [50]. Both adiponectin and its receptors (AdipoR1 and AdipoR2) are expressed in Leydig cells and seminiferous tubules; thus, the involvement of this hormone in testicular activities is reasonable [51].
Notably, conflicting results have been produced concerning the interplay between adiponectin and the HPT axis. Caminos et al. observed that treatment with adiponectin reduced GnRH-induced testosterone secretion in rat testes [52]. In contrast, when used at high doses, adiponectin stimulated the proliferation and survival of testicular cells, protected them from oxidative stress, and enhanced the transport of energetic substrates [53]. Compelling in vitro evidence also underlined that this adipokine may directly control the function of Leydig cells through the activation of key protein and enzymes involved in androgen synthesis (i.e., STAR, CYP11A1), as well as by promoting testosterone production in a cAMP–PKA-dependent manner [54,55]. Similar results were obtained in diabetic mice with increased body weight and reduced adiponectin levels, with testicular dysfunction improved after adiponectin replacement therapy in terms of upregulation of steroidogenesis, amelioration of insulin receptor-induced transport of energy substrates (glucose, lactate), and a marked reduction in oxidative stress markers in testes [56].
The role of adiponectin in the regulation of testicular function was also clarified by an in vivo model in which the genetic abrogation of AdipoR2 increased adiposity and caused atrophy of the seminiferous tubules and aspermia [57].
Considering these results, we can conclude that adiponectin is essential for the maintenance of the eugonadal state, as well as of normal reproductive activity. Therefore, abnormalities in its production and secretion, as typically observed in the setting of AT dysfunction, may exacerbate obesity-induced hypoandrogenemia.

2.3. Effects of Inflammation

Obesity is reportedly accompanied by low-grade inflammation. Under energy storage overload, an increased release of proinflammatory cytokines (i.e., TNF-α, IL-1β, IL-6), especially from the visceral AT, is well documented [58]. Accumulating evidence indicates that the overproduction of inflammatory mediators, besides leading to metabolic complications, is associated with reduced testosterone levels. Several studies have linked various acute and chronic inflammatory conditions (e.g., sepsis, burns, autoimmune diseases) to biochemical testosterone deficiency [59,60,61]. Large-scale epidemiological studies have associated obesity and systemic inflammation (e.g., elevated C-reactive protein (CRP)) with lower testosterone levels [62,63,64], whereas the administration of proinflammatory cytokines or endotoxins to men [65] or the treatment of Leydig cells in culture with proinflammatory cytokines have been shown to decrease testosterone production [66]. Taken together, these data provide strong evidence for a link between inflammation and testosterone deficiency.
Indeed, a negative correlation between IL-6 and testosterone concentrations was found in dysmetabolic men [67]. In a pro-inflammatory environment, a reduction in testosterone levels could occur through modifications of the HPT axis, as demonstrated in cultured anterior pituitary cells from male rats in which IL-6 suppressed GnRH-stimulated LH release [68]. Likewise, IL-1β and TNF-α were found to suppress the release of gonadotropin hormones, particularly LH, by interfering with the translational mechanisms of the GnRH transcript [69,70]. The noxious effect of inflammation on GnRH production is principally due to the reduced responsiveness of hypothalamic neurons to kisspeptins (Figure 1). The role of kisspeptin signaling in the maintenance of the balance of gonadotropins and androgens emerged from previous investigations where the infusion of exogenous intravenous kisspeptin-54 in healthy men stimulated a significant increase in serum LH, FSH, and testosterone concentrations [71]. However, in the presence of a proinflammatory condition, the activity of kisspeptin neurons is compromised, as reported by Sarchielli et al. who demonstrated that prolonged exposure to TNF-α inhibits gene expression of kisspeptin receptors (KISS1R) and affects the cilia volume of hypothalamic cells, thus interfering with the distribution of these receptors on the plasma membrane [72].
In detail, through the activation of TNF-α-induced Ikbeta kinase (IKK-beta)/NF-kB pathway, hypothalamic neurons lose the ability to secrete GnRH and develop a kisspeptin-resistance due to a ciliogenesis defect [72]. Corroborating in vivo results confirmed these data demonstrating the inhibitory effects of TNF-α on the GnRH–LH system and elucidating that C-reactive protein might also well interfere with the HPT axis, as well as participating in metabolic alterations [73].
Interestingly, as described before, several adipokines can directly suppress the HPT axis. However, numerous studies have clearly demonstrated how they can enhance (e.g., leptin or resistin) or alleviate (e.g., adiponectin and ghrelin), various features of inflammatory disease (e.g., asthma), acting either as pro- or anti-inflammatory factors, respectively [74,75]. Therefore, an indirect effect of adipokines mediated by inflammation may be also hypothesized.

2.4. Effect of Oxidative Stress

The expansion of AT requires the appropriate formation of new vessels to guarantee an adequate blood supply. Nevertheless, the overfeeding-induced increase in AT might occur at a rate difficult to be paralleled by a sufficient increase in blood flow and oxygen supply, thus leading to AT hypoxia [76]. Hypoxia may underpin oxidative stress in terms of generation of reactive oxygen species (ROS), providing additional routes by which low O2 tension can lead to adipose tissue dysfunction [77].
Several stressogenic stimuli are prominent in obesity, including hyperglycemia, hyperleptinemia, and mitochondrial failure, all known contributors of the increased generation of ROS and adipose tissue dysfunction [78,79,80].
Intense oxidative stress may affect the steroidogenic cascade in Leydig cells, eventually leading to decreased testosterone production and infertility. ROS can alter mitochondrial processes in Leydig cells by diminishing the expression of STAR, which can, in turn, decrease the mitochondrial transport of cholesterol and consequently reduce the synthesis of androgens [66,81,82].
In addition, the excessive production of ROS could participate in the onset of functional hypogonadism via an indirect mechanism, by enhancing cortisol secretion, which in turn affects LH secretion from pituitary gland, thus reducing testosterone production by Leydig cells [81] (Figure 1).
In this scenario, the ROS-dependent abrogation of testosterone secretion may also lead to a reduction in nitric oxide (NO) within the corpus cavernosum, thus impairing cavernous relaxation and favoring the development of erectile dysfunction [6,83]. Indeed, several studies have demonstrated that the restoration of the antioxidant system via either pharmacological compounds or genetic methods re-established erectile function through an increase in NO production and appeared to potentiate the efficacy of current pharmacological medications for sexual dysfunction [84,85,86]. Nevertheless, in the tunica albuginea compartment of the corpora cavernosa, ROS have been demonstrated to increase NO as well and its products of peroxidation (e.g., nitric oxide radical, peroxynitrite protonated, peroxynitrite nitrogen dioxide radical, nitronium ion) leading to chronic penile modifications, such as induratio penis plastica [87,88].

3. Effects of Pharmacological and Non-Pharmacological Approaches for Obesity on Hypogonadism

The mainstay of treatment for individuals with obesity is lifestyle interventions, such as hypocaloric diets. Nevertheless, notwithstanding their importance, it is undeniable that diet and behavioral therapies have quite an elevated rate of failure in the long-term, as demonstrated by the high rates of obesity relapse. For this reason, different weight-loss drugs have been developed, and several more are currently under investigation; each of them affects different biological and/or neuro-endocrine pathways, reflecting the complexity of energy balance and adipose tissue biology. Finally, bariatric surgery is indicated as the ultimate weight-loss strategy, since it not only reduces the amount of food ingested but also modifies the metabolic profile of the patient.
Many currently approved therapies for weight-loss have been recently investigated for their ability to also ameliorate obesity-induced hypogonadism. Herein, we discuss the current evidence on the effects of pharmacological and non-pharmacological therapies against hypogonadism and obesity on both the HPT axis and AT function.

3.1. Pharmacological Treatment

3.1.1. Orlistat

Orlistat, an inhibitor of intestinal lipase, promotes body weight reduction. However, body weight loss obtained with this drug is probably clinically irrelevant, even if statistically significant as compared to placebo (~−3.0 kg) [89]. Therapeutic effects of orlistat on obesity-induced steroidogenesis and spermatogenesis decline have been described in murine models. Indeed, treatment with orlistat has been associated with decreased leptin and increased adiponectin levels, improved sperm parameters, and decreased sperm DNA fragmentation; it also increased the levels of steroidogenic hormones, penile cGMP level, sexual behavior, and fertility outcome in obese male rats fed a high-fat diet [90,91]. However, orlistat treatment has not been proven to induce substantial effects on testosterone levels; thus, the potential recovery from functional hypogonadism in obesity is only speculative.

3.1.2. Glucagon-like Peptide-1 Receptor Agonists

Glucagon-like peptide-1 receptor agonists (GLP-1RA), an anti-diabetic drug class acting as incretin mimetic able to exert pleiotropic effects, represent one of the pharmacological approaches for the treatment of obesity [92,93,94,95].
Recently, the positive effects of GLP-1RA on both male fertility and AT health in obese men have been reported [6,96]. Indeed, studies of the GLP-1RA liraglutide and exenatide have suggested potential benefits when used as a co-therapy for andrological problems. In fact, a retrospective observational study reported that the supplementation of liraglutide to testosterone replacement therapy (TRT) allowed a consistent body weight reduction to be achieved and glycemic targets to be reached, together with a recovery in androgen levels in diabetic obese male subjects with overt hypogonadism [97]. Accordingly, short-term combined therapy with exenatide and metformin, a biguanide drug recommended as a first-line antidiabetic therapy, was found to favor the recovery of normal testosterone levels in association with the correction of sexual dysfunction in obese men with diabetes [98]. A preclinical study also found that liraglutide ameliorates hypogonadism symptoms together with improved glucose and lipid metabolism of AT when administered in orchiectomized rats [99]. Likewise, exenatide-based therapy mitigated the detrimental effects of obesity in HFD-fed male mice in terms of the amelioration of testicular inflammation, but did not yield significant improvements in testosterone levels [100].

3.2. Diet

Data from European Male Aging Study provide evidence that testosterone undergoes fluctuations by lifestyle factors according to the grade of weight change. In the setting of modest weight loss (<15%), a slight increase in total testosterone (+2 nmol/L) was observed, probably because of increases in SHBG levels, whereas free testosterone did not change [101]. However, with greater weight loss (>15%), total testosterone was definitely increased (+5.75 nmol/L), with significantly higher levels of free testosterone (+51.78 pmol/L), likely due to the activation of the HPT axis, as evidenced by a significant rise in LH release (+2 U/L) [101]. Nevertheless, the success of diet-induced weight-loss in the recovery of gonadal function is generally influenced by the short period of this intervention as compared with other clinical approaches (i.e., bariatric surgery). Indeed, a meal replacement-based low-calorie diet led to modest weight loss (−9.8%) with moderate increases in testosterone (2.9–5.1 nmol/L) as compared to surgical intervention, which was associated with a more robust weight-lowering effect (−32%) and testosterone restoration (7.8–12.5 nmol/L) [102,103,104,105].
Although previous findings have observed that dietary intervention leads to a weak modification of androgens levels, more recent studies in humans observed that the very low-calorie ketogenic diet (VLCKD) represents an effective tool against obesity and functional hypogonadism [106,107,108,109]. In particular, VLCKD treatment improves glucose homeostasis (83.5  ±  10.6 mg/dL), promotes weight loss (14.9 ± 3.9%), and restores β-cell secretory function in association with the improvement of gonadal functions and testosterone secretion (218.1  ±  53.9%) [107,108]. Therefore, even with the paucity of available data, VLCKD may be considered as an encouraging approach for obesity-related hypogonadism.

3.3. Bariatric Surgery

Despite the promising results of dietary intervention, bariatric surgery currently represents the best choice for the treatment of morbid obesity for rapid weight loss and metabolic improvement (i.e., reductions in glycaemia, insulinemia, triglycerides) [110].
Indeed, a recent meta-analysis illustrated that bariatric surgery is more effective in terms of weight-lowering efficacy (−32%) and testosterone recovery (+9 nmol/L) when compared to a low-calorie diet that induced smaller body weight reduction (−9.8%) and only a mild increase in testosterone (+3 nmol/L) [111]. Notably, not only was testosterone production affected by body weight modifications, but estrogen levels were also found to be reduced after bariatric surgery (−23 pmol/L), thus revealing an important adaptation of adipose tissue to weight loss. Importantly, high estrogen levels are known to inhibit kisspeptin expression in hypothalamic nuclei, and thus it is reasonable that the bariatric surgery-induced reduction in estrogens could improve the HPT axis function by upregulating kisspeptin expression in GnRH neurons [112].
Notably, the amelioration of testosterone concentrations after bariatric surgery appears to be correlated with the improvement in different parameters (e.g., body weight, BMI, adiponectin, leptin) regardless of the surgical technique (e.g., biliopancreatic diversion, vertical banded gastroplasty, Roux-en-Y gastric bypass) [113,114,115]. In addition, the loss of visceral AT could have a critical role in the post-surgical restoration of the eugonadal condition. Indeed, the reduction in VAT area assessed by MRI is positively associated with the increase in total testosterone plasma levels after bariatric surgery [116]. In addition to the contribution of weight loss per se, the restoration of normal testosterone levels after bariatric surgery could also be hypothesized to occur via the amelioration of inflammation markers since total testosterone levels were found to correlate significantly with changes in IL-6 and reductions in C-reactive protein [117].
Bariatric surgery appears to improve sexual health in obese men in terms of enhancement of sexual drive and erectile and ejaculatory function proportionately to the weight loss [118,119]. Particularly, after laparoscopic Roux-en-Y gastric bypass surgery, sperm volume and viability increased in association with decreased IL-8 and DNA fragmentation [120]. In contrast, previous findings observed that patients who underwent either gastric bypass surgery or sleeve gastrectomy had a reduction in total sperm count one-year after intervention [120], probably due to a rapid and sizeable excess body weight loss that led to a nutritional deficiency thus worsening the spermatogenesis [121]. Considering these results, surgical interventions produce successful and encouraging outcomes in terms of the patient’s overall health and restoration of eugonadal conditions, but are associated with conflicting data on sperm parameters. Thus, further studies on larger populations are needed to elucidate the effect on reproductive outcomes.

4. Effects of Testosterone Replacement Therapy on Adipose Tissue

One of the main therapeutic options for the treatment of functional hypogonadism is represented by testosterone replacement therapy (TRT) [122]. Interestingly, TRT might be an ally in promoting fat mass loss and improving metabolic outcomes through the direct actions of testosterone on AT function [10,123]. Indeed, the exogenous administration of testosterone undecanoate IM 1000 mg every 12 weeks was shown to lead to a significant decrease in body weight (~−15 kg), as well as waist circumference (~−12 cm) [124,125]. A meta-analysis by Corona et al. demonstrated that TRT in men with testosterone deficiency exerts its action mainly on the reduction in fat mass and the increase in lean mass, rather than on overall body weight [111]. Moreover, the loss of fat mass observed after TRT appears to relate particularly to the visceral compartment [106]. Interestingly, in castrated obese mice, testosterone-derived estradiol selectively blocks visceral fat growth by the reduction in adipocyte hypertrophy as well as the generation of new adipose cells [126].
The lipolytic efficacy of testosterone-based therapy is also accompanied by improvements in AT function. Indeed, nonhuman primates with androgen deficiency and dysfunctional white AT (i.e., multilocular and insulin-resistant adipocytes) restored normal AT functions and structure after TRT together with enhanced insulin sensitivity [127]. A recent cross-sectional analysis elucidated the mechanisms behind the anti-adipogenic ability of testosterone, observing that TRT enhanced the expression of genes involved in mitochondrial biogenesis and function (i.e., nrf1, tfam, fis1, etc.), lipid-handling genes (i.e., dio2, prkaca, and prkacb) and browning markers (pgc1α) in both visceral AT and preadipocytes from hypogonadal obese men [128].
However, when choosing this therapy, it must be kept in mind that even though TRT represents an appropriate strategy to reduce hypogonadal symptoms and adiposity, exogenous testosterone suppresses pituitary LH secretion and dampens spermatogenesis, exerting negative consequences on fertility [129].

5. Conclusions

In conclusion, dysfunctional AT plays a critical role in the development of functional hypogonadism in male obesity via both direct and indirect mechanisms. The dysregulation of adipokine secretion, inflammation, and oxidative stress promoted by fat mass expansion contribute, together with testosterone sequestration and inactivation mediated by AT, to the derangement of the HPT axis. On the other hand, hypogonadism fosters the expansion of AT, typically in the visceral compartment, mainly by lowering lipolytic rates and enhancing adipogenesis (Figure 1).
In this scenario, the reduction in fat mass obtained with pharmacological (i.e., orlistat and GLP-1RA) and non-pharmacological strategies (i.e., hypocaloric diet, VLCKD, bariatric surgery) has been shown to ameliorate testosterone levels and potentially male fertility. A remarkable exception is TRT, for which the metabolic improvement and weight loss attributable to exogenous testosterone is inevitably accompanied by infertility (Table 1).

Author Contributions

A.C. and V.A.G. contributed to conception of the review. A.C., V.A.G. and C.L. retrieved the main articles included in the review. A.C., V.A.G. and E.R. wrote the first draft of the manuscript. A.C. and F.G. supervised the project and finalized the manuscript. Review and editing, V.A.G., E.R., C.L. and R.D.; visualization, A.B., C.C. and G.P. All authors contributed to manuscript revision, read, and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This work was carried out with support from the ‘Fondazione per la Ricerca Biomedica Saverio e Isabella Cianciola’, Italy. The APC was funded by funds of the Specialization School in Endocrinology and Metabolic Diseases, University of Bari Aldo Moro.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abarca-Gómez, L.; Abdeen, Z.A.; Hamid, Z.A.; Abu-Rmeileh, N.M.; Acosta-Cazares, B.; Acuin, C.; Adams, R.J.; Aekplakorn, W.; Afsana, K.; Aguilar-Salinas, C.A.; et al. Worldwide Trends in Body-Mass Index, Underweight, Overweight, and Obesity from 1975 to 2016: A Pooled Analysis of 2416 Population-Based Measurement Studies in 1289 Million Children, Adolescents, and Adults. Lancet 2017, 390, 2627–2642. [Google Scholar] [CrossRef] [Green Version]
  2. Schoettl, T.; Fischer, I.P.; Ussar, S. Heterogeneity of Adipose Tissue in Development and Metabolic Function. J. Exp. Biol. 2018, 221 (Suppl. S1), jeb162958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Heid, I.M.; Jackson, A.U.; Randall, J.C.; Winkler, T.W.; Qi, L.; Steinthorsdottir, V.; Thorleifsson, G.; Zillikens, M.C.; Speliotes, E.K.; Mägi, R.; et al. Meta-Analysis Identifies 13 New Loci Associated with Waist-Hip Ratio and Reveals Sexual Dimorphism in the Genetic Basis of Fat Distribution. Nat. Genet. 2010, 42, 949–960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Randall, J.C.; Winkler, T.W.; Kutalik, Z.; Berndt, S.I.; Jackson, A.U.; Monda, K.L.; Kilpeläinen, T.O.; Esko, T.; Mägi, R.; Li, S.; et al. Sex-Stratified Genome-Wide Association Studies Including 270,000 Individuals Show Sexual Dimorphism in Genetic Loci for Anthropometric Traits. PLoS Genet. 2013, 9, e1003500. [Google Scholar] [CrossRef] [PubMed]
  5. Bhasin, S.; Cunningham, G.R.; Hayes, F.J.; Matsumoto, A.M.; Snyder, P.J.; Swerdloff, R.S.; Montori, V.M. Testosterone Therapy in Men with Androgen Deficiency Syndromes: An Endocrine Society Clinical Practice Guideline. J. Clin. Endocrinol. Metab. 2010, 95, 2536–2559. [Google Scholar] [CrossRef] [PubMed]
  6. Cignarelli, A.; Genchi, V.A.; D’Oria, R.; Giordano, F.; Caruso, I.; Perrini, S.; Natalicchio, A.; Laviola, L.; Giorgino, F. Role of Glucose-Lowering Medications in Erectile Dysfunction. J. Clin. Med. 2021, 10, 2501. [Google Scholar] [CrossRef] [PubMed]
  7. Niederberger, C. Re: Determinants of Testosterone Recovery after Bariatric Surgery: Is It Only a Matter of Reduction of Body Mass Index? J. Urol. 2014, 191, 753–754. [Google Scholar] [CrossRef]
  8. Kaplan, S.A.; Lee, J.Y.; O’Neill, E.A.; Meehan, A.G.; Kusek, J.W. Prevalence of Low Testosterone and Its Relationship to Body Mass Index in Older Men with Lower Urinary Tract Symptoms Associated with Benign Prostatic Hyperplasia. Aging Male 2013, 16, 169–172. [Google Scholar] [CrossRef]
  9. Molina-Vega, M.; Asenjo-Plaza, M.; García-Ruiz, M.C.; Varea-Marineto, E.; Casal-Nievas, N.; Álvarez-Millán, J.J.; Cabezas-Sanchez, P.; Cardona-Díaz, F.; Queipo-Ortuño, M.I.; Castellano-Castillo, D.; et al. Cross-Sectional, Primary Care-Based Study of the Prevalence of Hypoandrogenemia in Nondiabetic Young Men with Obesity. Obesity 2019, 27, 1584–1590. [Google Scholar] [CrossRef]
  10. Barone, B.; Napolitano, L.; Abate, M.; Cirillo, L.; Reccia, P.; Passaro, F.; Turco, C.; Morra, S.; Mastrangelo, F.; Scarpato, A.; et al. The Role of Testosterone in the Elderly: What Do We Know? Int. J. Mol. Sci. 2022, 23, 3535. [Google Scholar] [CrossRef]
  11. Carrageta, D.F.; Oliveira, P.F.; Alves, M.G.; Monteiro, M.P. Obesity and Male Hypogonadism: Tales of a Vicious Cycle. Obes. Rev. 2019, 20, 1148–1158. [Google Scholar] [CrossRef]
  12. Vermeulen, A.; Kaufman, J.M.; Deslypere, J.P.; Thomas, G. Attenuated Luteinizing Hormone (LH) Pulse Amplitude but Normal LH Pulse Frequency, and Its Relation to Plasma Androgens in Hypogonadism of Obese Men. J. Clin. Endocrinol. Metab. 1993, 76, 1140–1146. [Google Scholar] [CrossRef]
  13. Zumoff, B.; Miller, L.K.; Strain, G.W. Reversal of the Hypogonadotropic Hypogonadism of Obese Men by Administration of the Aromatase Inhibitor Testolactone. Metab. Clin. Exp. 2003, 52, 1126–1128. [Google Scholar] [CrossRef]
  14. Hofstra, J.; Loves, S.; van Wageningen, B.; Ruinemans-Koerts, J.; Jansen, I.; de Boer, H. High Prevalence of Hypogonadotropic Hypogonadism in Men Referred for Obesity Treatment. Neth. J. Med. 2008, 66, 103–109. [Google Scholar]
  15. Isidori, A.M.; Caprio, M.; Strollo, F.; Moretti, C.; Frajese, G.; Isidori, A.; Fabbri, A. Leptin and Androgens in Male Obesity: Evidence for Leptin Contribution to Reduced Androgen Levels. J. Clin. Endocrinol. Metab. 1999, 84, 3673–3680. [Google Scholar] [CrossRef] [Green Version]
  16. Di Vincenzo, A.; Busetto, L.; Vettor, R.; Rossato, M. Obesity, Male Reproductive Function and Bariatric Surgery. Front. Endocrinol. 2018, 9, 769. [Google Scholar] [CrossRef] [Green Version]
  17. Bélanger, C.; Luu-The, V.; Dupont, P.; Tchernof, A. Adipose Tissue Intracrinology: Potential Importance of Local Androgen/Estrogen Metabolism in the Regulation of Adiposity. Horm. Metab. Res. 2002, 34, 737–745. [Google Scholar] [CrossRef]
  18. Bélanger, C.; Hould, F.S.; Lebel, S.; Biron, S.; Brochu, G.; Tchernof, A. Omental and Subcutaneous Adipose Tissue Steroid Levels in Obese Men. Steroids 2006, 71, 674–682. [Google Scholar] [CrossRef]
  19. O’Reilly, M.W.; Kempegowda, P.; Walsh, M.; Taylor, A.E.; Manolopoulos, K.N.; Allwood, J.W.; Semple, R.K.; Hebenstreit, D.; Dunn, W.B.; Tomlinson, J.W.; et al. AKR1C3-Mediated Adipose Androgen Generation Drives Lipotoxicity in Women with Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab. 2017, 102, 3327–3339. [Google Scholar] [CrossRef]
  20. Di Nisio, A.; Sabovic, I.; De Toni, L.; Rocca, M.S.; Dall’Acqua, S.; Azzena, B.; De Rocco Ponce, M.; Foresta, C. Testosterone Is Sequestered in Dysfunctional Adipose Tissue, Modifying Androgen-Responsive Genes. Int. J. Obes. 2020, 44, 1617–1625. [Google Scholar] [CrossRef]
  21. Birzniece, V. Gonadal Steroids and Body Composition, Strength, and Sexual Function in Men. N. Engl. J. Med. 2013, 369, 2455. [Google Scholar] [CrossRef] [Green Version]
  22. Wake, D.J.; Strand, M.; Rask, E.; Westerbacka, J.; Livingstone, D.E.W.; Soderberg, S.; Andrew, R.; Yki-Jarvinen, H.; Olsson, T.; Walker, B.R. Intra-Adipose Sex Steroid Metabolism and Body Fat Distribution in Idiopathic Human Obesity. Clin. Endocrinol. 2007, 66, 440–446. [Google Scholar] [CrossRef]
  23. Xu, X.; Sun, M.; Ye, J.; Luo, D.; Su, X.; Zheng, D.; Feng, L.; Gao, L.; Yu, C.; Guan, Q. The Effect of Aromatase on the Reproductive Function of Obese Males. Horm. Metab. Res. 2017, 49, 572–579. [Google Scholar] [CrossRef]
  24. Biondi, G.; Marrano, N.; Borrelli, A.; Rella, M.; Palma, G.; Calderoni, I.; Siciliano, E.; Lops, P.; Giorgino, F.; Natalicchio, A. Adipose Tissue Secretion Pattern Influences β-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. Int. J. Mol. Sci. 2022, 23, 5522. [Google Scholar] [CrossRef]
  25. Perrini, S.; Cignarelli, A.; Quaranta, V.N.; Falcone, V.A.; Kounaki, S.; Porro, S.; Ciavarella, A.; Ficarella, R.; Barbaro, M.; Genchi, V.A.; et al. Correction of Intermittent Hypoxia Reduces Inflammation in Obese Subjects with Obstructive Sleep Apnea. JCI Insight 2017, 2, e94379. [Google Scholar] [CrossRef] [Green Version]
  26. Porro, S.; Genchi, V.A.; Cignarelli, A.; Natalicchio, A.; Laviola, L.; Giorgino, F.; Perrini, S. Dysmetabolic Adipose Tissue in Obesity: Morphological and Functional Characteristics of Adipose Stem Cells and Mature Adipocytes in Healthy and Unhealthy Obese Subjects. J. Endocrinol. Investig. 2020, 44, 921–941. [Google Scholar] [CrossRef]
  27. Genchi, V.A.; D’Oria, R.; Palma, G.; Caccioppoli, C.; Cignarelli, A.; Natalicchio, A.; Laviola, L.; Giorgino, F.; Perrini, S. Impaired Leptin Signalling in Obesity: Is Leptin a New Thermolipokine? Int. J. Mol. Sci. 2021, 22, 6445. [Google Scholar] [CrossRef]
  28. Aquila, S.; Gentile, M.; Middea, E.; Catalano, S.; Morelli, C.; Pezzi, V.; Andò, S. Leptin Secretion by Human Ejaculated Spermatozoa. J. Clin. Endocrinol. Metab. 2005, 90, 4753–4761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Leisegang, K.; Bouic, P.J.D.; Menkveld, R.; Henkel, R.R. Obesity Is Associated with Increased Seminal Insulin and Leptin alongside Reduced Fertility Parameters in a Controlled Male Cohort. Reprod. Biol. Endocrinol. RBE 2014, 12, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Fernandes Negris Lima, T.; Nackeeran, S.; Rakitina, E.; Fernandes Negris Lima, G.; Arora, H.; Kargi, A.Y.; Ramasamy, R. Association of Leptin with Total and Free Testosterone: Results from the National Health and Nutrition Examination Surveys. Androg. Clin. Res. Ther. 2020, 1, 94–100. [Google Scholar] [CrossRef] [PubMed]
  31. Einollahi, N.; Dashti, N.; Emamgholipour, S.; Zarebavani, M.; Sedighi-Gilani, M.A.; Choobineh, H. Evidence for Alteration in Serum Concentrations of Leptin in Infertile Men Categorized Based on BMI. Clin. Lab. 2016, 62, 2361–2366. [Google Scholar] [CrossRef]
  32. Smith, J.T.; Acohido, B.V.; Clifton, D.K.; Steiner, R.A. KiSS-1 Neurones Are Direct Targets for Leptin in the Ob/Ob Mouse. J. Neuroendocrinol. 2006, 18, 298–303. [Google Scholar] [CrossRef]
  33. Smith, J.T.; Cunningham, M.J.; Rissman, E.F.; Clifton, D.K.; Steiner, R.A. Regulation of Kiss1 Gene Expression in the Brain of the Female Mouse. Endocrinology 2005, 146, 3686–3692. [Google Scholar] [CrossRef]
  34. Roseweir, A.K.; Kauffman, A.S.; Smith, J.T.; Guerriero, K.A.; Morgan, K.; Pielecka-Fortuna, J.; Pineda, R.; Gottsch, M.L.; Tena-Sempere, M.; Moenter, S.M.; et al. Discovery of Potent Kisspeptin Antagonists Delineate Physiological Mechanisms of Gonadotropin Regulation. J. Neurosci. Off. J. Soc. Neurosci. 2009, 29, 3920–3929. [Google Scholar] [CrossRef]
  35. True, C.; Kirigiti, M.; Ciofi, P.; Grove, K.L.; Smith, M.S. Characterisation of Arcuate Nucleus Kisspeptin/Neurokinin B Neuronal Projections and Regulation during Lactation in the Rat. J. Neuroendocrinol. 2011, 23, 52–64. [Google Scholar] [CrossRef] [Green Version]
  36. Tena-Sempere, M.; Barreiro, M.L. Leptin in Male Reproduction: The Testis Paradigm. Mol. Cell. Endocrinol. 2002, 188, 9–13. [Google Scholar] [CrossRef]
  37. Ishikawa, T.; Fujioka, H.; Ishimura, T.; Takenaka, A.; Fujisawa, M. Expression of Leptin and Leptin Receptor in the Testis of Fertile and Infertile Patients. Andrologia 2007, 39, 22–27. [Google Scholar] [CrossRef]
  38. Myers, M.G. Leptin Receptor Signaling and the Regulation of Mammalian Physiology. Recent Prog. Horm. Res. 2004, 59, 287–304. [Google Scholar] [CrossRef]
  39. Kloek, C.; Haq, A.K.; Dunn, S.L.; Lavery, H.J.; Banks, A.S.; Myers, M.G. Regulation of Jak Kinases by Intracellular Leptin Receptor Sequences. J. Biol. Chem. 2002, 277, 41547–41555. [Google Scholar] [CrossRef] [Green Version]
  40. Banks, A.S.; Davis, S.M.; Bates, S.H.; Myers, M.G. Activation of Downstream Signals by the Long Form of the Leptin Receptor. J. Biol. Chem. 2000, 275, 14563–14572. [Google Scholar] [CrossRef] [Green Version]
  41. Bjørbæk, C.; Lavery, H.J.; Bates, S.H.; Olson, R.K.; Davis, S.M.; Flier, J.S.; Myers, M.G. SOCS3 Mediates Feedback Inhibition of the Leptin Receptor via Tyr985. J. Biol. Chem. 2000, 275, 40649–40657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Roumaud, P.; Martin, L.J. Roles of Leptin, Adiponectin and Resistin in the Transcriptional Regulation of Steroidogenic Genes Contributing to Decreased Leydig Cells Function in Obesity. Horm. Mol. Biol. Clin. Investig. 2015, 24, 25–45. [Google Scholar] [CrossRef] [PubMed]
  43. Yuan, M.; Huang, G.; Li, J.; Zhang, J.; Li, F.; Li, K.; Gao, B.; Zeng, L.; Shan, W.; Lin, P.; et al. Hyperleptinemia Directly Affects Testicular Maturation at Different Sexual Stages in Mice, and Suppressor of Cytokine Signaling 3 Is Involved in This Process. Reprod. Biol. Endocrinol. RBE 2014, 12, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhao, J.; Zhai, L.; Liu, Z.; Wu, S.; Xu, L. Leptin Level and Oxidative Stress Contribute to Obesity-Induced Low Testosterone in Murine Testicular Tissue. Oxid. Med. Cell. Longev. 2014, 2014, 190945. [Google Scholar] [CrossRef]
  45. Landry, D.A.; Sormany, F.; Haché, J.; Roumaud, P.; Martin, L.J. Steroidogenic Genes Expressions Are Repressed by High Levels of Leptin and the JAK/STAT Signaling Pathway in MA-10 Leydig Cells. Mol. Cell. Biochem. 2017, 433, 79–95. [Google Scholar] [CrossRef]
  46. Gao, Q.; Wolfgang, M.J.; Neschen, S.; Morino, K.; Horvath, T.L.; Shulman, G.I.; Fu, X.Y. Disruption of Neural Signal Transducer and Activator of Transcription 3 Causes Obesity, Diabetes, Infertility, and Thermal Dysregulation. Proc. Natl. Acad. Sci. USA 2004, 101, 4661–4666. [Google Scholar] [CrossRef] [Green Version]
  47. Cignarelli, A.; Genchi, V.A.; Perrini, S.; Natalicchio, A.; Laviola, L.; Giorgino, F. Insulin and Insulin Receptors in Adipose Tissue Development. Int. J. Mol. Sci. 2019, 20, 759. [Google Scholar] [CrossRef] [Green Version]
  48. Hoffstedt, J.; Arvidsson, E.; Sjölin, E.; Wåhlén, K.; Arner, P. Adipose Tissue Adiponectin Production and Adiponectin Serum Concentration in Human Obesity and Insulin Resistance. J. Clin. Endocrinol. Metab. 2004, 89, 1391–1396. [Google Scholar] [CrossRef] [Green Version]
  49. Neeland, I.J.; Ayers, C.R.; Rohatgi, A.K.; Turer, A.T.; Berry, J.D.; Das, S.R.; Vega, G.L.; Khera, A.; McGuire, D.K.; Grundy, S.M.; et al. Associations of Visceral and Abdominal Subcutaneous Adipose Tissue with Markers of Cardiac and Metabolic Risk in Obese Adults. Obesity 2013, 21, E439–E447. [Google Scholar] [CrossRef] [Green Version]
  50. Bai, J.; Liu, Y.; Niu, G.F.; Bai, L.X.; Xu, X.Y.; Zhang, G.Z.; Wang, L.X. Relationship between Adiponectin and Testosterone in Patients with Type 2 Diabetes. Biochem. Med. 2011, 21, 65–70. [Google Scholar] [CrossRef] [Green Version]
  51. Nogueiras, R.; Barreiro, M.L.; Caminos, J.E.; Gaytán, F.; Suominen, J.S.; Navarro, V.M.; Casanueva, F.F.; Aguilar, E.; Toppari, J.; Diéguez, C.; et al. Novel Expression of Resistin in Rat Testis: Functional Role and Regulation by Nutritional Status and Hormonal Factors. J. Cell Sci. 2004, 117, 3247–3257. [Google Scholar] [CrossRef] [Green Version]
  52. Caminos, J.E.; Nogueiras, R.; Gaytán, F.; Pineda, R.; González, C.R.; Barreiro, M.L.; Castaño, J.P.; Malagón, M.M.; Pinilla, L.; Toppari, J.; et al. Novel Expression and Direct Effects of Adiponectin in the Rat Testis. Endocrinology 2008, 149, 3390–3402. [Google Scholar] [CrossRef] [Green Version]
  53. Choubey, M.; Ranjan, A.; Bora, P.S.; Baltazar, F.; Krishna, A. Direct Actions of Adiponectin on Changes in Reproductive, Metabolic, and Anti-Oxidative Enzymes Status in the Testis of Adult Mice. Gen. Comp. Endocrinol. 2019, 279, 1–11. [Google Scholar] [CrossRef] [Green Version]
  54. Landry, D.; Paré, A.; Jean, S.; Martin, L.J. Adiponectin Influences Progesterone Production from MA-10 Leydig Cells in a Dose-Dependent Manner. Endocrine 2015, 48, 957–967. [Google Scholar] [CrossRef]
  55. Otani, M.; Kogo, M.; Furukawa, S.; Wakisaka, S.; Maeda, T. The Adiponectin Paralog C1q/TNF-Related Protein 3 (CTRP3) Stimulates Testosterone Production through the CAMP/PKA Signaling Pathway. Cytokine 2012, 58, 238–244. [Google Scholar] [CrossRef]
  56. Choubey, M.; Ranjan, A.; Bora, P.S.; Krishna, A. Protective Role of Adiponectin against Testicular Impairment in High-Fat Diet/Streptozotocin-Induced Type 2 Diabetic Mice. Biochimie 2020, 168, 41–52. [Google Scholar] [CrossRef]
  57. Bjursell, M.; Ahnmark, A.; Bohlooly-Y, M.; William-Olsson, L.; Rhedin, M.; Peng, X.R.; Ploj, K.; Gerdin, A.K.; Arnerup, G.; Elmgren, A.; et al. Opposing Effects of Adiponectin Receptors 1 and 2 on Energy Metabolism. Diabetes 2007, 56, 583–593. [Google Scholar] [CrossRef] [Green Version]
  58. Kawai, T.; Autieri, M.V.; Scalia, R. Adipose Tissue Inflammation and Metabolic Dysfunction in Obesity. Am. J. Physiol. Cell Physiol. 2021, 320, C375–C391. [Google Scholar] [CrossRef]
  59. Ballinger, A.B.; Savage, M.O.; Sanderson, I.R. Delayed Puberty Associated with Inflammatory Bowel Disease. Pediatr. Res 2003, 53, 205–210. [Google Scholar] [CrossRef]
  60. Christeff, N.; Benassayag, C.; Carli-Vielle, C.; Carli, A.; Nunez, E.A. Elevated Oestrogen and Reduced Testosterone Levels in the Serum of Male Septic Shock Patients. J. Steroid Biochem. 1988, 29, 435–440. [Google Scholar] [CrossRef]
  61. Lephart, E.D.; Baxter, C.R.; Parker, C.R. Effect of Burn Trauma on Adrenal and Testicular Steroid Hormone Production. J. Clin. Endocrinol. Metab. 1987, 64, 842–848. [Google Scholar] [CrossRef]
  62. Yeap, B.B.; Knuiman, M.W.; Divitini, M.L.; Handelsman, D.J.; Beilby, J.P.; Beilin, J.; McQuillan, B.; Hung, J. Differential Associations of Testosterone, Dihydrotestosterone and Oestradiol with Physical, Metabolic and Health-Related Factors in Community-Dwelling Men Aged 17-97 Years from the Busselton Health Survey. Clin. Endocrinol. 2014, 81, 100–108. [Google Scholar] [CrossRef]
  63. Tsilidis, K.K.; Rohrmann, S.; McGlynn, K.A.; Nyante, S.J.; Lopez, D.S.; Bradwin, G.; Feinleib, M.; Joshu, C.E.; Kanarek, N.; Nelson, W.G.; et al. Association between Endogenous Sex Steroid Hormones and Inflammatory Biomarkers in US Men. Andrology 2013, 1, 919–928. [Google Scholar] [CrossRef] [Green Version]
  64. Bobjer, J.; Katrinaki, M.; Tsatsanis, C.; Lundberg Giwercman, Y.; Giwercman, A. Negative Association between Testosterone Concentration and Inflammatory Markers in Young Men: A Nested Cross-Sectional Study. PLoS ONE 2013, 8, e61466. [Google Scholar] [CrossRef] [Green Version]
  65. Tremellen, K.; McPhee, N.; Pearce, K.; Benson, S.; Schedlowski, M.; Engler, H. Endotoxin-Initiated Inflammation Reduces Testosterone Production in Men of Reproductive Age. Am. J. Physiol.-Endocrinol. Metab. 2018, 314, E206–E213. [Google Scholar] [CrossRef] [Green Version]
  66. Hales, D.B. Testicular Macrophage Modulation of Leydig Cell Steroidogenesis. J. Reprod. Immunol. 2002, 57, 3–18. [Google Scholar] [CrossRef]
  67. Gautier, A.; Bonnet, F.; Dubois, S.; Massart, C.; Grosheny, C.; Bachelot, A.; Aubé, C.; Balkau, B.; Ducluzeau, P.-H. Associations between Visceral Adipose Tissue, Inflammation and Sex Steroid Concentrations in Men. Clin. Endocrinol. 2013, 78, 373–378. [Google Scholar] [CrossRef]
  68. Russell, S.H.; Small, C.J.; Stanley, S.A.; Franks, S.; Ghatei, M.A.; Bloom, S.R. The in Vitro Role of Tumour Necrosis Factor-Alpha and Interleukin-6 in the Hypothalamic-Pituitary Gonadal Axis. J. Neuroendocrinol. 2001, 13, 296–301. [Google Scholar] [CrossRef]
  69. Laaksonen, D.; Niskanen, L.; Punnonen, K.; Nyyssonen, K.; Tuomainen, T.; Salonen, R.; Rauramaa, R.; Salonen, J. Sex Hormones, Inflammation and the Metabolic Syndrome: A Population-Based Study. Eur. J. Endocrinol. 2003, 149, 601–608. [Google Scholar] [CrossRef]
  70. Watanobe, H.; Hayakawa, Y. Hypothalamic Interleukin-1β and Tumor Necrosis Factor-α, But Not Interleukin-6, Mediate the Endotoxin-Induced Suppression of the Reproductive Axis in Rats. Endocrinology 2003, 144, 4868–4875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Dhillo, W.S.; Chaudhri, O.B.; Patterson, M.; Thompson, E.L.; Murphy, K.G.; Badman, M.K.; McGowan, B.M.; Amber, V.; Patel, S.; Ghatei, M.A.; et al. Kisspeptin-54 Stimulates the Hypothalamic-Pituitary Gonadal Axis in Human Males. J. Clin. Endocrinol. Metab. 2005, 90, 6609–6615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sarchielli, E.; Comeglio, P.; Squecco, R.; Ballerini, L.; Mello, T.; Guarnieri, G.; Idrizaj, E.; Mazzanti, B.; Vignozzi, L.; Gallina, P.; et al. Tumor Necrosis Factor α Impairs Kisspeptin Signaling in Human Gonadotropin-Releasing Hormone Primary Neurons. J. Clin. Endocrinol. Metab. 2016, 102, 46–56. [Google Scholar] [CrossRef] [PubMed]
  73. Bhatia, V.; Chaudhuri, A.; Tomar, R.; Dhindsa, S.; Ghanim, H.; Dandona, P. Low Testosterone and High C-Reactive Protein Concentrations Predict Low Hematocrit in Type 2 Diabetes. Diabetes Care 2006, 29, 2289–2294. [Google Scholar] [CrossRef] [Green Version]
  74. Kaczyńska, K.; Zając, D.; Wojciechowski, P.; Jampolska, M. Regulatory Peptides in Asthma. Int. J. Mol. Sci. 2021, 22, 13656. [Google Scholar] [CrossRef]
  75. Miethe, S.; Guarino, M.; Alhamdan, F.; Simon, H.-U.; Renz, H.; Dufour, J.-F.; Potaczek, D.P.; Garn, H. Effects of Obesity on Asthma: Immunometabolic Links. Pol. Arch. Intern. Med. 2018, 128, 469–477. [Google Scholar] [CrossRef] [Green Version]
  76. Engin, A. Adipose Tissue Hypoxia in Obesity and Its Impact on Preadipocytes and Macrophages: Hypoxia Hypothesis. Adv. Exp. Med. Biol. 2017, 960, 305–326. [Google Scholar] [CrossRef]
  77. Carrière, A.; Carmona, M.-C.; Fernandez, Y.; Rigoulet, M.; Wenger, R.H.; Pénicaud, L.; Casteilla, L. Mitochondrial Reactive Oxygen Species Control the Transcription Factor CHOP-10/GADD153 and Adipocyte Differentiation: A Mechanism for Hypoxia-Dependent Effect. J. Biol. Chem. 2004, 279, 40462–40469. [Google Scholar] [CrossRef] [Green Version]
  78. Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef] [Green Version]
  79. Be\ltowski, J.; Wójcicka, G.; Jamroz, A. Leptin Decreases Plasma Paraoxonase 1 (PON1) Activity and Induces Oxidative Stress: The Possible Novel Mechanism for Proatherogenic Effect of Chronic Hyperleptinemia. Atherosclerosis 2003, 170, 21–29. [Google Scholar] [CrossRef]
  80. Fortuño, A.; Bidegain, J.; Baltanás, A.; Moreno, M.U.; Montero, L.; Landecho, M.F.; Beloqui, O.; Díez, J.; Zalba, G. Is Leptin Involved in Phagocytic NADPH Oxidase Overactivity in Obesity? Potential Clinical Implications. J. Hypertens. 2010, 28, 1944–1950. [Google Scholar] [CrossRef]
  81. Roychoudhury, S.; Chakraborty, S.; Choudhury, A.P.; Das, A.; Jha, N.K.; Slama, P.; Nath, M.; Massanyi, P.; Ruokolainen, J.; Kesari, K.K. Environmental Factors-Induced Oxidative Stress: Hormonal and Molecular Pathway Disruptions in Hypogonadism and Erectile Dysfunction. Antioxidants 2021, 10, 837. [Google Scholar] [CrossRef] [PubMed]
  82. Tsai, S.-C.; Lu, C.-C.; Lin, C.-S.; Wang, P.S. Antisteroidogenic Actions of Hydrogen Peroxide on Rat Leydig Cells. J. Cell. Biochem. 2003, 90, 1276–1286. [Google Scholar] [CrossRef] [PubMed]
  83. Cartledge, J.; Minhas, S.; Eardley, I. The Role of Nitric Oxide in Penile Erection. Expert Opin. Pharmacother. 2001, 2, 95–107. [Google Scholar] [CrossRef] [PubMed]
  84. Goswami, S.K.; Gangadarappa, S.K.; Vishwanath, M.; Razdan, R.; Jamwal, R.; Bhadri, N.; Inamdar, M.N. Antioxidant Potential and Ability of Phloroglucinol to Decrease Formation of Advanced Glycation End Products Increase Efficacy of Sildenafil in Diabetes-Induced Sexual Dysfunction of Rats. Sex. Med. 2016, 4, e104–e112. [Google Scholar] [CrossRef] [Green Version]
  85. Deng, W.; Bivalacqua, T.J.; Champion, H.C.; Hellstrom, W.J.; Murthy, S.N.; Kadowitz, P.J. Superoxide Dismutase—A Target for Gene Therapeutic Approach to Reduce Oxidative Stress in Erectile Dysfunction. Methods Mol. Biol. 2009, 610, 213–227. [Google Scholar] [CrossRef]
  86. Angulo, J.; Peiró, C.; Cuevas, P.; Gabancho, S.; Fernández, A.; González-Corrochano, R.; La Fuente, J.M.; Baron, A.D.; Chen, K.S.; De Tejada, I.S. The Novel Antioxidant, AC3056 (2,6-Di-t-Butyl-4-((Dimethyl-4-Methoxyphenylsilyl)Methyloxy)Phenol), Reverses Erectile Dysfunction in Diabetic Rats and Improves NO-Mediated Responses in Penile Tissue from Diabetic Men. J. Sex. Med. 2009, 6, 373–387. [Google Scholar] [CrossRef]
  87. Crocetto, F.; Barone, B.; Manfredi, C.; Trama, F.; Romano, L.; Romeo, M.; Russo, G.; Sicignano, E.; Persico, F.; Aveta, A.; et al. Are Insulin Resistance and Non-Alcoholic Fatty Liver Disease Associated with Peyronie’s Disease? A Pilot Study. J. Physiol. Pharm. 2022, 73, 1. [Google Scholar] [CrossRef]
  88. Paulis, G.; Brancato, T. Inflammatory Mechanisms and Oxidative Stress in Peyronies Disease: Therapeutic “Rationale” and Related Emerging Treatment Strategies. Inflamm. Allergy-Drug Targets 2012, 11, 48–57. [Google Scholar] [CrossRef] [Green Version]
  89. Sahebkar, A.; Simental-Mendía, L.E.; Reiner, Ž.; Kovanen, P.T.; Simental-Mendía, M.; Bianconi, V.; Pirro, M. Effect of Orlistat on Plasma Lipids and Body Weight: A Systematic Review and Meta-Analysis of 33 Randomized Controlled Trials. Pharmacol. Res. 2017, 122, 53–65. [Google Scholar] [CrossRef]
  90. Suleiman, J.B.; Nna, V.U.; Othman, Z.A.; Zakaria, Z.; Bakar, A.B.A.; Mohamed, M. Orlistat Attenuates Obesity-Induced Decline in Steroidogenesis and Spermatogenesis by up-Regulating Steroidogenic Genes. Andrology 2020, 8, 1471–1485. [Google Scholar] [CrossRef]
  91. Suleiman, J.B.; Nna, V.U.; Zakaria, Z.; Othman, Z.A.; Bakar, A.B.A.; Usman, U.Z.; Mohamed, M. Orlistat Reverses Intratesticular Lactate Transport Decline and Infertility in Male Obese Rats. Reproduction 2020, 160, 863–872. [Google Scholar] [CrossRef]
  92. Marrano, N.; Biondi, G.; Cignarelli, A.; Perrini, S.; Laviola, L.; Giorgino, F.; Natalicchio, A. Functional Loss of Pancreatic Islets in Type 2 Diabetes: How Can We Halt It? Metab. Clin. Exp. 2020, 110, 154304. [Google Scholar] [CrossRef]
  93. Santilli, F.; Simeone, P.G.; Guagnano, M.T.; Leo, M.; Maccarone, M.T.; Castelnuovo, A.D.; Sborgia, C.; Bonadonna, R.C.; Angelucci, E.; Federico, V.; et al. Effects of Liraglutide on Weight Loss, Fat Distribution, and β-Cell Function in Obese Subjects With Prediabetes or Early Type 2 Diabetes. Diabetes Care 2017, 40, 1556–1564. [Google Scholar] [CrossRef] [Green Version]
  94. Potts, J.E.; Gray, L.J.; Brady, E.M.; Khunti, K.; Davies, M.J.; Bodicoat, D.H. The Effect of Glucagon-Like Peptide 1 Receptor Agonists on Weight Loss in Type 2 Diabetes: A Systematic Review and Mixed Treatment Comparison Meta-Analysis. PLoS ONE 2015, 10, e0126769. [Google Scholar] [CrossRef] [Green Version]
  95. Halawi, H.; Khemani, D.; Eckert, D.; O’Neill, J.; Kadouh, H.; Grothe, K.; Clark, M.M.; Burton, D.D.; Vella, A.; Acosta, A.; et al. Effects of Liraglutide on Weight, Satiation, and Gastric Functions in Obesity: A Randomised, Placebo-Controlled Pilot Trial. Lancet Gastroenterol. Hepatol. 2017, 2, 890–899. [Google Scholar] [CrossRef]
  96. Cannarella, R.; Calogero, A.E.; Condorelli, R.A.; Greco, E.A.; Aversa, A.; La Vignera, S. Is There a Role for Glucagon-like Peptide-1 Receptor Agonists in the Treatment of Male Infertility? Andrology 2021, 9, 1499–1503. [Google Scholar] [CrossRef]
  97. Giagulli, V.A.; Carbone, M.D.; Ramunni, M.I.; Licchelli, B.; De Pergola, G.; Sabbà, C.; Guastamacchia, E.; Triggiani, V. Adding Liraglutide to Lifestyle Changes, Metformin and Testosterone Therapy Boosts Erectile Function in Diabetic Obese Men with Overt Hypogonadism. Andrology 2015, 3, 1094–1103. [Google Scholar] [CrossRef]
  98. Shao, N.; Yu, X.-Y.; Yu, Y.-M.; Li, B.-W.; Pan, J.; Wu, W.-H.; Zhang, H.-J.; Ma, X.-F.; Hao, M.; Kuang, H.-Y. Short-Term Combined Treatment with Exenatide and Metformin Is Superior to Glimepiride Combined Metformin in Improvement of Serum Testosterone Levels in Type 2 Diabetic Patients with Obesity. Andrologia 2018, 50, e13039. [Google Scholar] [CrossRef]
  99. Model, J.F.A.; Lima, M.V.; Ohlweiler, R.; Sarapio, E.; Vogt, É.L.; Rocha, D.S.; de Souza, S.K.; Vinagre, A.S. Liraglutide Treatment Counteracts Alterations in Adipose Tissue Metabolism Induced by Orchiectomy in Rats. Life Sci. 2021, 278, 119586. [Google Scholar] [CrossRef]
  100. Zhang, E.; Xu, F.; Liang, H.; Yan, J.; Xu, H.; Li, Z.; Wen, X.; Weng, J. GLP-1 Receptor Agonist Exenatide Attenuates the Detrimental Effects of Obesity on Inflammatory Profile in Testis and Sperm Quality in Mice. Am. J. Reprod. Immunol. 2015, 74, 457–466. [Google Scholar] [CrossRef]
  101. Camacho, E.M.; Huhtaniemi, I.T.; O’Neill, T.W.; Finn, J.D.; Pye, S.R.; Lee, D.M.; Tajar, A.; Bartfai, G.; Boonen, S.; Casanueva, F.F.; et al. Age-Associated Changes in Hypothalamic-Pituitary-Testicular Function in Middle-Aged and Older Men Are Modified by Weight Change and Lifestyle Factors: Longitudinal Results from the European Male Ageing Study. Eur. J. Endocrinol. 2013, 168, 445–455. [Google Scholar] [CrossRef]
  102. Corona, G.; Rastrelli, G.; Monami, M.; Saad, F.; Luconi, M.; Lucchese, M.; Facchiano, E.; Sforza, A.; Forti, G.; Mannucci, E.; et al. Body Weight Loss Reverts Obesity-Associated Hypogonadotropic Hypogonadism: A Systematic Review and Meta-Analysis. Eur. J. Endocrinol. 2013, 168, 829–843. [Google Scholar] [CrossRef] [Green Version]
  103. Reis, L.O.; Favaro, W.J.; Barreiro, G.C.; de Oliveira, L.C.; Chaim, E.A.; Fregonesi, A.; Ferreira, U. Erectile Dysfunction and Hormonal Imbalance in Morbidly Obese Male Is Reversed after Gastric Bypass Surgery: A Prospective Randomized Controlled Trial. Int. J. Androl. 2010, 33, 736–744. [Google Scholar] [CrossRef]
  104. Niskanen, L.; Laaksonen, D.E.; Punnonen, K.; Mustajoki, P.; Kaukua, J.; Rissanen, A. Changes in Sex Hormone-Binding Globulin and Testosterone during Weight Loss and Weight Maintenance in Abdominally Obese Men with the Metabolic Syndrome. Diabetes Obes. Metab. 2004, 6, 208–215. [Google Scholar] [CrossRef]
  105. Kaukua, J.; Pekkarinen, T.; Sane, T.; Mustajoki, P. Sex Hormones and Sexual Function in Obese Men Losing Weight. Obes. Res. 2003, 11, 689–694. [Google Scholar] [CrossRef]
  106. Caliber, M.; Hackett, G. Important Lessons about Testosterone Therapy- Weight Loss vs. Testosterone Therapy for Symptom Resolution, Classical vs. Functional Hypogonadism, and Shortterm vs. Lifelong Testosterone Therapy. Aging Male Off. J. Int. Soc. Study Aging Male 2020, 23, 585–591. [Google Scholar] [CrossRef]
  107. La Vignera, S.; Cannarella, R.; Galvano, F.; Grillo, A.; Aversa, A.; Cimino, L.; Magagnini, C.M.; Mongioì, L.M.; Condorelli, R.A.; Calogero, A.E. The Ketogenic Diet Corrects Metabolic Hypogonadism and Preserves Pancreatic SS-Cell Function in Overweight/Obese Men: A Single-Arm Uncontrolled Study. Endocrine 2021, 72, 392–399. [Google Scholar] [CrossRef]
  108. Mongioì, L.M.; Cimino, L.; Condorelli, R.A.; Magagnini, M.C.; Barbagallo, F.; Cannarella, R.; Vignera, S.L.; Calogero, A.E. Effectiveness of a Very Low Calorie Ketogenic Diet on Testicular Function in Overweight/Obese Men. Nutrients 2020, 12, 2967. [Google Scholar] [CrossRef]
  109. Mongioì, L.M.; Cimino, L.; Greco, E.; Cannarella, R.; Condorelli, R.A.; La Vignera, S.; Calogero, A.E. Very-Low-Calorie Ketogenic Diet: An Alternative to a Pharmacological Approach to Improve Glycometabolic and Gonadal Profile in Men with Obesity. Curr. Opin. Pharmacol. 2021, 60, 72–82. [Google Scholar] [CrossRef]
  110. Capoccia, D.; Coccia, F.; Guarisco, G.; Testa, M.; Rendina, R.; Abbatini, F.; Silecchia, G.; Leonetti, F. Long-Term Metabolic Effects of Laparoscopic Sleeve Gastrectomy. Obes. Surg. 2018, 28, 2289–2296. [Google Scholar] [CrossRef]
  111. Corona, G.; Vignozzi, L.; Sforza, A.; Mannucci, E.; Maggi, M. Obesity and Late-Onset Hypogonadism. Mol. Cell. Endocrinol. 2015, 418 Pt 2, 120–133. [Google Scholar] [CrossRef] [PubMed]
  112. Morelli, A.; Marini, M.; Mancina, R.; Luconi, M.; Vignozzi, L.; Fibbi, B.; Filippi, S.; Pezzatini, A.; Forti, G.; Vannelli, G.B.; et al. Sex Steroids and Leptin Regulate the “First Kiss” (KiSS 1/G-Protein-Coupled Receptor 54 System) in Human Gonadotropin-Releasing-Hormone-Secreting Neuroblasts. J. Sex. Med. 2008, 5, 1097–1113. [Google Scholar] [CrossRef] [PubMed]
  113. Hammoud, A.; Gibson, M.; Hunt, S.C.; Adams, T.D.; Carrell, D.T.; Kolotkin, R.L.; Meikle, A.W. Effect of Roux-En-Y Gastric Bypass Surgery on the Sex Steroids and Quality of Life in Obese Men. J. Clin. Endocrinol. Metab. 2009, 94, 1329–1332. [Google Scholar] [CrossRef] [PubMed]
  114. 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. [Google Scholar] [CrossRef] [Green Version]
  115. Calderón, B.; Galdón, A.; Calañas, A.; Peromingo, R.; Galindo, J.; García-Moreno, F.; Rodriguez-Velasco, G.; Martín-Hidalgo, A.; Vazquez, C.; Escobar-Morreale, H.F.; et al. Effects of Bariatric Surgery on Male Obesity-Associated Secondary Hypogonadism: Comparison of Laparoscopic Gastric Bypass with Restrictive Procedures. Obes. Surg. 2014, 24, 1686–1692. [Google Scholar] [CrossRef]
  116. Liu, F.; Tu, Y.; Zhang, P.; Bao, Y.; Han, J.; Jia, W. Decreased Visceral Fat Area Correlates with Improved Total Testosterone Levels after Roux-En-Y Gastric Bypass in Obese Chinese Males with Type 2 Diabetes: A 12-Month Follow-Up. Surg. Obes. Relat. Dis. 2018, 14, 462–468. [Google Scholar] [CrossRef]
  117. Zhu, C.; Mei, F.; Gao, J.; Zhou, D.; Lu, L.; Qu, S. Changes in Inflammatory Markers Correlated with Increased Testosterone after Laparoscopic Sleeve Gastrectomy in Obese Chinese Men with Acanthosis Nigricans. J. Dermatol. 2019, 46, 338–342. [Google Scholar] [CrossRef]
  118. Glina, F.P.A.; de Freitas Barboza, J.W.; Nunes, V.M.; Glina, S.; Bernardo, W.M. What Is the Impact of Bariatric Surgery on Erectile Function? A Systematic Review and Meta-Analysis. Sex. Med. Rev. 2017, 5, 393–402. [Google Scholar] [CrossRef]
  119. Sarhan, M.D.; Khattab, M.; Sarhan, M.D.; Maurice, K.K.; Hassan, H. Impact of Bariatric Surgery on Male Sexual Health: A Prospective Study. Obes. Surg. 2021, 31, 4064–4069. [Google Scholar] [CrossRef]
  120. Kun, L.; Pin, Z.; Jianzhong, D.; Xiaodong, H.; Haoyong, Y.; Yuqian, B.; Hongwei, Z. Significant Improvement of Erectile Function after Roux-En-Y Gastric Bypass Surgery in Obese Chinese Men with Erectile Dysfunction. Obes. Surg. 2015, 25, 838–844. [Google Scholar] [CrossRef]
  121. Carette, C.; Levy, R.; Eustache, F.; Baron, G.; Coupaye, M.; Msika, S.; Barrat, C.; Cohen, R.; Catheline, J.M.; Brugnon, F.; et al. Changes in Total Sperm Count after Gastric Bypass and Sleeve Gastrectomy: The BARIASPERM Prospective Study. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2019, 15, 1271–1279. [Google Scholar] [CrossRef]
  122. Bhasin, S.; Brito, J.P.; Cunningham, G.R.; Hayes, F.J.; Hodis, H.N.; Matsumoto, A.M.; Snyder, P.J.; Swerdloff, R.S.; Wu, F.C.; Yialamas, M.A. Testosterone Therapy in Men with Hypogonadism: An Endocrine Society* Clinical Practice Guideline. J. Clin. Endocrinol. Metab. 2018, 103, 1715–1744. [Google Scholar] [CrossRef] [Green Version]
  123. Grossmann, M.; Tang Fui, M.; Dupuis, P. Lowered Testosterone in Male Obesity: Mechanisms, Morbidity and Management. Asian J. Androl. 2014, 16, 223. [Google Scholar] [CrossRef]
  124. Saad, F.; Aversa, A.; Isidori, A.M.; Gooren, L.J. Testosterone as Potential Effective Therapy in Treatment of Obesity in Men with Testosterone Deficiency: A Review. Curr. Diabetes Rev. 2012, 8, 131–143. [Google Scholar] [CrossRef] [Green Version]
  125. Haider, A.; Yassin, A.; Doros, G.; Saad, F. Effects of Long-Term Testosterone Therapy on Patients with “Diabesity”: Results of Observational Studies of Pooled Analyses in Obese Hypogonadal Men with Type 2 Diabetes. Int. J. Endocrinol. 2014, 2014, 683515. [Google Scholar] [CrossRef]
  126. Sebo, Z.L.; Rodeheffer, M.S. Testosterone Metabolites Differentially Regulate Obesogenesis and Fat Distribution. Mol. Metab. 2021, 44, 101141. [Google Scholar] [CrossRef]
  127. Varlamov, O.; White, A.E.; Carroll, J.M.; Bethea, C.L.; Reddy, A.; Slayden, O.; O’Rourke, R.W.; Roberts, C.T. Androgen Effects on Adipose Tissue Architecture and Function in Nonhuman Primates. Endocrinology 2012, 153, 3100–3110. [Google Scholar] [CrossRef] [Green Version]
  128. Maseroli, E.; Comeglio, P.; Corno, C.; Cellai, I.; Filippi, S.; Mello, T.; Galli, A.; Rapizzi, E.; Presenti, L.; Truglia, M.C.; et al. Testosterone Treatment Is Associated with Reduced Adipose Tissue Dysfunction and Nonalcoholic Fatty Liver Disease in Obese Hypogonadal Men. J. Endocrinol. Investig. 2021, 44, 819–842. [Google Scholar] [CrossRef]
  129. Page, S.T.; Kalhorn, T.F.; Bremner, W.J.; Anawalt, B.D.; Matsumoto, A.M.; Amory, J.K. Intratesticular Androgens and Spermatogenesis during Severe Gonadotropin Suppression Induced by Male Hormonal Contraceptive Treatment. J. Androl. 2007, 28, 734–741. [Google Scholar] [CrossRef]
Figure 1. The vicious cycle of obesity–hypogonadism. Obesity-induced AT dysfunction impaired the secretion of several mediators affecting the release of GnRH from the hypothalamus as well as of FSH and LH from the pituitary gland. Low levels of these hormones worsen testicular function in terms of the reduction of testosterone production, leading to overt hypogonadism. The decrease in testosterone levels further exacerbates the impairment of AT in terms of a decreased rate of lipolysis and increased lipogenesis, the extent of which accelerates the failure of HPT axis, thus sustaining a feedforward detrimental loop. Abbreviations: adiponectin receptor (AdipoR); follicle-stimulating hormone (FSH); gonadotropin-releasing hormone (GnRH); interleukin-1β (IL-1β); interleukin-6 (IL-6); suppressor of cytokine signaling 3 (SOCS3); tumor necrosis factor α (TNFα); leptin receptor (LEPR); luteinizing hormone (LH); LH receptor (LHR); phosphorylated Janus kinase 2 (pJAK2); reactive oxygen species (ROS); steroidogenic acute regulatory protein (STAR); signal transducer and activator of transcriptor 3 (STAT3).
Figure 1. The vicious cycle of obesity–hypogonadism. Obesity-induced AT dysfunction impaired the secretion of several mediators affecting the release of GnRH from the hypothalamus as well as of FSH and LH from the pituitary gland. Low levels of these hormones worsen testicular function in terms of the reduction of testosterone production, leading to overt hypogonadism. The decrease in testosterone levels further exacerbates the impairment of AT in terms of a decreased rate of lipolysis and increased lipogenesis, the extent of which accelerates the failure of HPT axis, thus sustaining a feedforward detrimental loop. Abbreviations: adiponectin receptor (AdipoR); follicle-stimulating hormone (FSH); gonadotropin-releasing hormone (GnRH); interleukin-1β (IL-1β); interleukin-6 (IL-6); suppressor of cytokine signaling 3 (SOCS3); tumor necrosis factor α (TNFα); leptin receptor (LEPR); luteinizing hormone (LH); LH receptor (LHR); phosphorylated Janus kinase 2 (pJAK2); reactive oxygen species (ROS); steroidogenic acute regulatory protein (STAR); signal transducer and activator of transcriptor 3 (STAT3).
Ijms 23 08194 g001
Table 1. Effects of different approaches for obesity treatment and for hypogonadal hypogonadism on testosterone levels and fertility.
Table 1. Effects of different approaches for obesity treatment and for hypogonadal hypogonadism on testosterone levels and fertility.
InterventionBody WeightTFertility
ORLISTAT-↑ (?)
GLP-1RA↓↓-
VLCKD↓↓-
BARIATRIC SURGERY↓↓↓↑↑+/−
TRT↑↑
↑ increase; ↓ decrease; - not available; +/− conflicting results; ? uncertain results; ↔ unchanged.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Genchi, V.A.; Rossi, E.; Lauriola, C.; D’Oria, R.; Palma, G.; Borrelli, A.; Caccioppoli, C.; Giorgino, F.; Cignarelli, A. Adipose Tissue Dysfunction and Obesity-Related Male Hypogonadism. Int. J. Mol. Sci. 2022, 23, 8194. https://doi.org/10.3390/ijms23158194

AMA Style

Genchi VA, Rossi E, Lauriola C, D’Oria R, Palma G, Borrelli A, Caccioppoli C, Giorgino F, Cignarelli A. Adipose Tissue Dysfunction and Obesity-Related Male Hypogonadism. International Journal of Molecular Sciences. 2022; 23(15):8194. https://doi.org/10.3390/ijms23158194

Chicago/Turabian Style

Genchi, Valentina Annamaria, Erica Rossi, Celeste Lauriola, Rossella D’Oria, Giuseppe Palma, Anna Borrelli, Cristina Caccioppoli, Francesco Giorgino, and Angelo Cignarelli. 2022. "Adipose Tissue Dysfunction and Obesity-Related Male Hypogonadism" International Journal of Molecular Sciences 23, no. 15: 8194. https://doi.org/10.3390/ijms23158194

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop