scispace - formally typeset
Search or ask a question

Showing papers on "Testosterone published in 2015"


Journal ArticleDOI
TL;DR: The sex-based differences in skeletal muscle fiber composition, myosin heavy chain expression, contractile function, and the regulation by thyroid hormone, estrogen, and testosterone are reviewed.
Abstract: Previous studies have identified over 3,000 genes that are differentially expressed in male and female skeletal muscle. Here, we review the sex-based differences in skeletal muscle fiber composition, myosin heavy chain expression, contractile function, and the regulation of these physiological differences by thyroid hormone, estrogen, and testosterone. The findings presented lay the basis for the continued work needed to fully understand the skeletal muscle differences between males and females.

256 citations


Journal ArticleDOI
TL;DR: Intermittent treatment with high-dose testosterone shows therapeutic potential in patients with castration-resistant prostate cancer and may restore the tumors’ sensitivity to antiandrogen agents, further expanding patients’ treatment options.
Abstract: Targeting androgen receptor (AR) axis signaling by disrupting androgen-AR interactions remains the primary treatment for metastatic prostate cancer. Unfortunately, all men develop resistance to primary castrating therapy and secondary androgen deprivation therapies (ADTs). Resistance develops in part because castration-resistant prostate cancer (CRPC) cells adaptively up-regulate AR levels through overexpression, amplification, and expression of ligand-independent variants in response to chronic exposure to a low-testosterone environment. However, preclinical models suggest that AR overexpression represents a therapeutic liability that can be exploited via exposure to supraphysiologic testosterone to promote CRPC cell death. Preclinical data supported a pilot study in which 16 asymptomatic CRPC patients with low to moderate metastatic burden were treated with testosterone cypionate (400 mg intramuscular; day 1 of 28) and etoposide (100 mg oral daily; days 1 to 14 of 28). After three cycles, those with a declining prostate-specific antigen (PSA) continued on intermittent testosterone therapy monotherapy. Castrating therapy was continued to suppress endogenous testosterone production, allowing for rapid cycling from supraphysiologic to near-castrate serum testosterone levels, a strategy termed bipolar androgen therapy (BAT). BAT was well tolerated and resulted in high rates of PSA (7 of 14 evaluable patients) and radiographic responses (5 of 10 evaluable patients). Although all men showed eventual PSA progression, four men remained on BAT for ≥1 year. All patients (10 of 10) demonstrated PSA reductions upon receiving androgen-ablative therapies after BAT, suggesting that BAT may also restore sensitivity to ADTs. BAT shows promise as treatment for CRPC and should be further evaluated in larger trials.

210 citations


Journal ArticleDOI
TL;DR: It is demonstrated for the first time that adult male exposure to BPA caused a reduction in sperm production and specific functional parameters, and the corresponding pattern of gene expression is indicative of an attempt by the pituitary to reestablish normal levels of LH, FSH and testosterone serum concentrations.

185 citations


Journal ArticleDOI
TL;DR: These models have pinpointed the steps of spermatogenesis that require AR signalling and detail the essential nature of androgens in the promotion of male fertility.

171 citations


Journal ArticleDOI
TL;DR: In transgender women, estrogen therapy, with or without antiandrogen therapy, was associated with lower BP, and in transgender men, testosterone therapy wasassociated with increased BMI.

149 citations


Journal ArticleDOI
TL;DR: Stress as a causative factor in male infertility cannot be ignored and patients should be made aware of its effects on testicular function and fertility and helped to manage them.
Abstract: Psychological stress can be defined as any uncomfortable 'emotional experience' accompanied by predictable biochemical, physiological and behavioural changes or responses. Many clinical studies looking at the effects of psychological stress on male fertility have shown that stress is associated with reduced paternity and abnormal semen parameters. Enough scientific evidence exists to suggest that psychological stress could severely affect spermatogenesis, mainly as a result of varying testosterone secretion. The hypothalamic-pituitary-adrenal axis has a direct inhibitory action on the hypothalamic-pituitary-gonadal (HPG) axis and Leydig cells in the testes. The newly discovered hormone, gonadotropin-inhibitory hormone (GnIH), also has an inhibitory effect on the HPG axis. Inhibition of the HPG axis results in a fall in testosterone levels, which causes changes in Sertoli cells and the blood-testis barrier, leading to the arrest of spermatogenesis. Germ cells also become vulnerable to gonadotoxins and oxidation. However, the extent and severity of the effects of psychological stress on human testes is difficult to study and data mostly come from animal models. Despite this limitation, stress as a causative factor in male infertility cannot be ignored and patients should be made aware of its effects on testicular function and fertility and helped to manage them.

139 citations


Journal ArticleDOI
TL;DR: Low nadir serum testosterone within the first year of androgen-deprivation therapy correlates with improved CSS and duration of response to androgen deprivation in men being treated for biochemical failure undergoing CAD.
Abstract: Purpose Three small retrospective studies have suggested that patients undergoing continuous androgen deprivation (CAD) have superior survival and time to progression if lower castrate levels of testosterone ( 0.7 nmol/L) are achieved. Evidence from prospective large studies has been lacking. Patients and Methods The PR-7 study randomly assigned patients experiencing biochemical failure after radiation therapy or surgery plus radiation therapy to CAD or intermittent androgen deprivation. The relationship between testosterone levels in the first year and cause-specific survival (CSS) and time to androgen-independent progression in men in the CAD arm was evaluated using Cox regression. Results There was a significant difference in CSS (P .015) and time to hormone resistance (P .02) among those who had first-year minimum nadir testosterone 0.7, 0.7 to 1.7, and 1.7 nmol/L. Patients with first-year nadir testosterone consistently 0.7 nmol/L had significantly higher risks of dying as a result of disease (0.7 to 1.7 nmol/L: hazard ratio [HR], 2.08; 95% CI, 1.28 to 3.38; 1.7 nmol/L: HR, 2.93; 95% CI, 0.70 to 12.30) and developing hormone resistance (0.7 to 1.7 nmol/L: HR, 1.62; 95% CI, 1.20 to 2.18; 1.7 nmol/L: HR, 1.90; 95% CI, 0.77 to 4.70). Maximum testosterone 1.7 nmol/L predicted for a higher risk of dying as a result of disease (P .02).

137 citations


Journal ArticleDOI
Yong Fan1, Yue Liu1, Ke Xue1, Guobao Gu, Weimin Fan1, Yali Xu1, Zhide Ding1 
17 Apr 2015-PLOS ONE
TL;DR: Obesity can impair male fertility through declines in the sperm function parameters, sex hormone level, whereas during spermatogenesis damage to the blood-testis barrier (BTB) integrity may be one of the crucial underlying factors accounting for this change.
Abstract: Obesity is a complex metabolic disease that is a serious detriment to both children and adult health, which induces a variety of diseases, such as cardiovascular disease, type II diabetes, hypertension and cancer. Although adverse effects of obesity on female reproduction or oocyte development have been well recognized, its harmfulness to male fertility is still unclear because of reported conflicting results. The aim of this study was to determine whether diet-induced obesity impairs male fertility and furthermore to uncover its underlying mechanisms. Thus, male C57BL/6 mice fed a high-fat diet (HFD) for 10 weeks served as a model of diet-induced obesity. The results clearly show that the percentage of sperm motility and progressive motility significantly decreased, whereas the proportion of teratozoospermia dramatically increased in HFD mice compared to those in normal diet fed controls. Besides, the sperm acrosome reaction fell accompanied by a decline in testosterone level and an increase in estradiol level in the HFD group. This alteration of sperm function parameters strongly indicated that the fertility of HFD mice was indeed impaired, which was also validated by a low pregnancy rate in their mated normal female. Moreover, testicular morphological analyses revealed that seminiferous epithelia were severely atrophic, and cell adhesions between spermatogenic cells and Sertoli cells were loosely arranged in HFD mice. Meanwhile, the integrity of the blood-testis barrier was severely interrupted consistent with declines in the tight junction related proteins, occludin, ZO-1 and androgen receptor, but instead endocytic vesicle-associated protein, clathrin rose. Taken together, obesity can impair male fertility through declines in the sperm function parameters, sex hormone level, whereas during spermatogenesis damage to the blood-testis barrier (BTB) integrity may be one of the crucial underlying factors accounting for this change.

136 citations


Journal ArticleDOI
TL;DR: A short overview of the possible actions of dehydroepiandrosterone and its sulfate in the brain is presented, calling attention to various mechanisms of their action as neurosteroids and to prospects for the knowledge of their role in brain disorders.

112 citations


Journal ArticleDOI
TL;DR: The results suggest that protracted use of acetaminophen (1 week) may suppress fetal testosterone production, which could have adverse consequences, and further studies are required to establish the dose-response and treatment-duration relationships.
Abstract: Most common male reproductive disorders are linked to lower testosterone exposure in fetal life, although the factors responsible for suppressing fetal testosterone remain largely unknown. Protracted use of acetaminophen during pregnancy is associated with increased risk of cryptorchidism in sons, but effects on fetal testosterone production have not been demonstrated. We used a validated xenograft model to expose human fetal testes to clinically relevant doses and regimens of acetaminophen. Exposure to a therapeutic dose of acetaminophen for 7 days significantly reduced plasma testosterone (45% reduction; P = 0.025) and seminal vesicle weight (a biomarker of androgen exposure; 18% reduction; P = 0.005) in castrate host mice bearing human fetal testis xenografts, whereas acetaminophen exposure for just 1 day did not alter either parameter. Plasma acetaminophen concentrations (at 1 hour after the final dose) in exposed host mice were substantially below those reported in humans after a therapeutic oral dose. Subsequent in utero exposure studies in rats indicated that the acetaminophen-induced reduction in testosterone likely results from reduced expression of key steroidogenic enzymes (Cyp11a1, Cyp17a1). Our results suggest that protracted use of acetaminophen (1 week) may suppress fetal testosterone production, which could have adverse consequences. Further studies are required to establish the dose-response and treatment-duration relationships to delineate the maximum dose and treatment period without this adverse effect.

106 citations


Journal ArticleDOI
TL;DR: The known hormonal changes and their possible regulators in on-time and premature adrenarche are described, and the clinical features and possible later health problems associating with PA are described.

Journal ArticleDOI
TL;DR: This study explores the possible association between the use of two typical 5ARIs (finasteride and dutasteride) and the subsequent risk of osteoporosis diagnosis.
Abstract: SummaryBackground 5-alpha-reductase inhibitors (5ARIs) are the potent androgen responsible for the development and enlargement of the prostate gland by decreasing dihydrotestosterone (DHT). This results in inhibition of the conversion of testosterone to dihydrotestosterone and markedly suppresses serum dihydrotestosterone levels. Testosterone replacement therapy improves bone density in men with hypogonadal osteoporosis. This study explores the possible association between the use of two typical 5ARIs (finasteride and dutasteride) and the subsequent risk of osteoporosis diagnosis. Methods We identified 1352 osteoporosis diagnosis cases and 5387 control cases without osteoporosis diagnosis from the claims data for patients with benign prostate hyperplasia (BPH), which are collected in the Taiwanese National Health Insurance Research Database (NHIRD). Four controls were frequency matched to each case according to age (every 5 years) and diagnosis date. We measured the effect of 5ARIs and determined the adjusted odds ratios (ORs) with 95% confidence intervals (CIs). Results We observed a 1·52-fold increase in osteoporosis diagnosis among patients with BPH using finasteride (95% CI, 1·01–2·30). Furthermore, a dosage analysis showed that higher doses of finasteride were associated with higher osteoporosis diagnosis risk (OR = 1·68; 95% CI, 1·01–2·81), relative to the patients not using 5ARIs. Conclusion This population-based nested case–control study suggests that the use of finasteride can increase the risk of osteoporosis diagnosis among patients with BPH. The effects were more prominent in patients using higher doses of finasteride.

Journal ArticleDOI
TL;DR: The results indicate that the TRPM8 channel is physically associated with testosterone and suggest that, in addition to a genomic role, testosterone plays a role in direct regulation of the TR PM8 channel function.

Journal ArticleDOI
TL;DR: Evidence for disordered androgen receptor function in the amygdala is described, together with changes in estrogen receptor-α, serotonergic and GABAergic genes in thegdala, and hippocampus, which suggests that maternal androgen excess may underpin the risk of developing anxiety disorders in daughters and sons of PCOS mothers.
Abstract: During pregnancy, women with polycystic ovary syndrome (PCOS) display high circulating androgen levels that may affect the fetus and increase the risk of mood disorders in offspring. This study investigated whether maternal androgen excess causes anxiety-like behavior in offspring mimicking anxiety disorders in PCOS. The PCOS phenotype was induced in rats following prenatal androgen (PNA) exposure. PNA offspring displayed anxiety-like behavior in the elevated plus maze, which was reversed by flutamide [androgen receptor (AR) blocker] and tamoxifen [selective estrogen receptor (ER) modulator]. Circulating sex steroids did not differ between groups at adult age. The expression of serotonergic and GABAergic genes associated with emotional regulation in the amygdala was consistent with anxiety-like behavior in female, and partly in male PNA offspring. Furthermore, AR expression in amygdala was reduced in female PNA offspring and also in females exposed to testosterone in adult age. To determine whether AR activation in amygdala affects anxiety-like behavior, female rats were given testosterone microinjections into amygdala, which resulted in anxiety-like behavior. Together, these data describe the anxiety-like behavior in PNA offspring and adult females with androgen excess, an impact that seems to occur during fetal life, and is mediated via AR in amygdala, together with changes in ERα, serotonergic, and GABAergic genes in amygdala and hippocampus. The anxiety-like behavior following testosterone microinjections into amygdala demonstrates a key role for AR activation in this brain area. These results suggest that maternal androgen excess may underpin the risk of developing anxiety disorders in daughters and sons of PCOS mothers.

Journal ArticleDOI
TL;DR: There are promising new approaches to increase serum T by directly stimulating Leydig cell T production rather than by exogenous T therapy, thus potentially avoiding some of its negative consequences.

Book ChapterDOI
01 Jan 2015
TL;DR: Whereas metabolic information provides permissive signals that time sexual maturity in both sexes, the presence or absence of prenatal programming by testosterone determines whether photoperiod also serves as a permissive cue to begin reproductive activity in sheep.
Abstract: In sheep, internal and external cues are integrated to time the decrease in sensitivity to steroid negative feedback that increases the frequency of gonadotropin-releasing hormone (GnRH) pulses to drive the transition into adulthood. The neural substrate that processes the information used to time the pubertal increase in GnRH secretion is programmed prenatally by the organizational actions of testosterone produced in the male, but not the female (the default sex), in response to an early increase in GnRH and gonadotropin secretion. In both sexes, metabolic cues provide information that sufficient growth has occurred to begin reproduction. However, the young female, being inherently photoperiodic, cannot express her sexual maturity until day length is appropriate. Once she has experienced the long days of summer followed by the short days of autumn, neuroendocrine sensitivity to steroid negative feedback decreases, and high-frequency GnRH pulses are expressed to initiate ovarian cycles. By contrast, prenatal testicular steroids masculinize the photoneuroendocrine system to reduce the reliance on day length as a cue for puberty, and the male lamb does not use photoperiods to time the expression of high-frequency GnRH pulses. Once metabolic signals indicate that energy balance is sufficient in the male, high-frequency GnRH pulses are expressed. Thus, whereas metabolic information provides permissive signals that time sexual maturity in both sexes, the presence or absence of prenatal programming by testosterone determines whether photoperiod also serves as a permissive cue to begin reproductive activity. Viewed in a broad sense, the neuroendocrine response to internal and external signals timing the transition to adulthood has fetal origins in the sheep.

Journal ArticleDOI
TL;DR: Evidence is provided for a strong link between a high TT/DHT ratio and an adverse metabolic phenotype in PCOS patients and significant correlations of this ratio with various adverse anthropometric, hormonal, lipid and liver parameters and parameters of glucose metabolism were found.
Abstract: Context: Polycystic ovary syndrome (PCOS) is a heterogeneous disease with many different aspects, including hyperandrogenism and metabolic disturbances. Clinical phenotypes show different patterns of steroid hormones that have been investigated to some extent. Objective: This study intended to determine the role of the testosterone (TT) to dihydrotestosterone (DHT) ratio (TT/DHT ratio) in PCOS patients and to further assess the correlation of this ratio with hormonal, anthropometric, and metabolic parameters. Design and Setting: Serum samples of 275 premenopausal PCOS patients fulfilling Rotterdam criteria and 35 BMI-matched, premenopausal, healthy controls were analyzed for testosterone, DHT, dehydroepiandrosterone (DHEA), and androstenedione using liquid chromatography/mass spectrometry. Main Outcome Measures: We measured total levels of testosterone and DHT and calculated unbound hormone levels as well as the ratio of testosterone to DHT. Further, impaired glucose tolerance, basal and stimulated serum ...

Journal ArticleDOI
TL;DR: The use of TRT in prostate cancer patients is still considered experimental and should only be offered after well-informed shared decision making and with close monitoring, as there is no clear evidence that elevations in endogenous testosterone levels promote the development of prostate cancer in humans.
Abstract: Testosterone plays a central role in male development and health. Likewise, androgen deficiency, or hypogonadism, is associated with a variety of symptoms including decreased energy, diminished libido and erectile dysfunction, among others. Male androgen levels steadily decline with age, and, in a subset of symptomatic older men, can result in late-onset hypogonadism (LOH). Over the last decade, increased awareness of hypogonadism among patients and providers has led to a significant rise in the use of testosterone replacement therapy (TRT) for hypogonadism, and especially in LOH. Accompanying the rise in TRT are concerns of potential adverse effects, including cardiovascular risks and the promotion of prostate cancer. The 'androgen hypothesis' asserts that prostate cancer development and progression is driven by androgens, and thus TRT has the theoretical potential to drive prostate cancer development and progression. In this review, we examine existing data surrounding testosterone and prostate cancer. There is significant evidence that androgens promote prostate cancer in experimental systems. However, there is no clear evidence that elevations in endogenous testosterone levels promote the development of prostate cancer in humans. As a result of experimental and historical data on the progression of prostate cancer following TRT, there has been widespread belief that TRT will promote disease progression in prostate cancer patients. Despite these fears, there are a growing number of studies demonstrating no increase in prostate cancer incidence among men on TRT. Furthermore, in studies involving a small number of patients, there has been no discernable increase in disease progression in prostate cancer patients on TRT. While data from large, prospective, randomized, controlled trials are absent, TRT in select prostate cancer patients is likely safe. In the end, the use of TRT in prostate cancer patients is still considered experimental and should only be offered after well-informed shared decision making and with close monitoring.

Journal ArticleDOI
TL;DR: It was difficult to predict which hepatic drug-metabolizing P450 or steroidogenic P450 will be mainly responsible for metabolizing each steroid hormone in vivo based on these results, and further research is required.
Abstract: This article reviews in vitro metabolic activities [including Michaelis constants (Km), maximal velocities (Vmax) and Vmax/Km] and drug-steroid interactions [such as induction and cooperativity (activation)] of cytochromes P450 (P450 or CYP) in human tissues, including liver and adrenal gland, for 14 kinds of endogenous steroid compounds, including allopregnanolone, cholesterol, cortisol, cortisone, dehydroepiandrosterone, estradiol, estrone, pregnenolone, progesterone, testosterone and bile acids (cholic acid). First, we considered the drug-metabolizing P450s. 6β-Hydroxylation of many steroids, including cortisol, cortisone, progesterone and testosterone, was catalyzed primarily by CYP3A4. CYP1A2 and CYP3A4, respectively, are likely the major hepatic enzymes responsible for 2-/4-hydroxylation and 16α-hydroxylation of estradiol and estrone, steroids that can contribute to breast cancer risk. In contrast, CYP1A1 and CYP1B1 predominantly metabolized estrone and estradiol to 2- and 4-catechol estrogens, which are endogenous ultimate carcinogens if formed in the breast. Some metabolic activities of CYP3A4, including dehydroepiandrosterone 7β-/16α-hydroxylation, estrone 2-hydroxylation and testosterone 6β-hydroxylation, were higher than those for polymorphically expressed CYP3A5. Next, we considered typical steroidogenic P450s. CYP17A1, CYP19A1 and CYP27A1 catalyzed steroid synthesis, including hydroxylation at 17α, 19 and 27 positions, respectively. However, it was difficult to predict which hepatic drug-metabolizing P450 or steroidogenic P450 will be mainly responsible for metabolizing each steroid hormone in vivo based on these results. Further research is required on the metabolism of steroid hormones by various P450s and on prediction of their relative contributions to in vivo metabolism. The findings collected here provide fundamental and useful information on the metabolism of steroid compounds.

Journal ArticleDOI
TL;DR: It is concluded that autocrine androgen action within Leydig cells is essential for the lifelong support of spermatogenesis and the development and lifelong health of Leydigs cells.
Abstract: Leydig cell number and function decline as men age, and low testosterone is associated with all "Western" cardio-metabolic disorders. However, whether perturbed androgen action within the adult Leydig cell lineage predisposes individuals to this late-onset degeneration remains unknown. To address this, we generated a novel mouse model in which androgen receptor (AR) is ablated from ∼75% of adult Leydig stem cell/cell progenitors, from fetal life onward (Leydig cell AR knockout mice), permitting interrogation of the specific roles of autocrine Leydig cell AR signaling through comparison to adjacent AR-retaining Leydig cells, testes from littermate controls, and to human testes, including from patients with complete androgen insensitivity syndrome (CAIS). This revealed that autocrine AR signaling is dispensable for the attainment of final Leydig cell number but is essential for Leydig cell maturation and regulation of steroidogenic enzymes in adulthood. Furthermore, these studies reveal that autocrine AR signaling in Leydig cells protects against late-onset degeneration of the seminiferous epithelium in mice and inhibits Leydig cell apoptosis in both adult mice and patients with CAIS, possibly via opposing aberrant estrogen signaling. We conclude that autocrine androgen action within Leydig cells is essential for the lifelong support of spermatogenesis and the development and lifelong health of Leydig cells.

Journal ArticleDOI
TL;DR: Stress affects the behavioral, endocrine and the molecular response of the stress systems in the hypothalamus of SD rats in a clear sexual dimorphic way, which has parallels in human data on stress and depression.

Journal ArticleDOI
TL;DR: The chromatin-immunoprecipitation analysis of TRPM8 found that an androgen response element (ARE) mediates androgen regulation of trpm8 and indicates that the testosterone-induced PMTRPM8 activity elicits Ca2+ uptake, subsequently causing apoptotic cell death.
Abstract: // Swapna Asuthkar 1 , Kiran Kumar Velpula 1 , Pia A. Elustondo 2 , Lusine Demirkhanyan 1 and Eleonora Zakharian 1 1 University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA 2 Dalhousie University, Halifax, NS, Canada Correspondence to: Eleonora Zakharian, email: // Swapna Asuthkar, email: // Keywords : prostate cancer (PC), transient receptor potential melastatin 8(TRPM8) ion channel, testosterone, DHT (5α-dihydrotestosterone), androgen receptor (AR) Received : March 18, 2015 Accepted : April 07, 2015 Published : April 29, 2015 Abstract The cold and menthol receptor TRPM8 is highly expressed in prostate and prostate cancer (PC). Recently, we identified that TRPM8 is as an ionotropic testosterone receptor. The TRPM8 mRNA is expressed in early prostate tumors with high androgen levels, while anti-androgen therapy greatly reduces its expression. Here, from the chromatin-immunoprecipitation (ChIP) analysis, we found that an androgen response element (ARE) mediates androgen regulation of trpm8 . Furthermore, using immunofluorescence, calcium-imaging and planar lipid bilayers, we identified that TRPM8 channel is functionally regulated by androgens in the prostate. Although TRPM8 mRNA is expressed at high levels, we found that the TRPM8 protein undergoes ubiquitination and degradation in PC cells. The mass-spectrometry analysis of TRPM8, immunoprecipitated from LNCaP cells identified ubiquitin-like modifier-activating enzyme 1 (UBA1). PYR-41, a potent inhibitor of initial enzyme in the ubiquitination cascade, UBA1, increased TRPM8 activity on the plasma membrane (PM) of LNCaP cells. Furthermore, PYR-41-mediated PM TRPM8 activity was accompanied by enhanced activation of p53 and Caspase-9. Interestingly, we found that the trpm8 promoter possesses putative binding sites for p53 and that the overexpression of p53 increased the TRPM8 mRNA levels. In addition to the genomic regulation of TRPM8 by AR and p53, our findings indicate that the testosterone-induced PM TRPM8 activity elicits Ca 2+ uptake, subsequently causing apoptotic cell death. These findings support the strategy of rescuing PM TRPM8 expression as a new therapeutic application through the regulation of PC cell growth and proliferation.

Journal ArticleDOI
TL;DR: The cross-sex steroid hormone treatment of transsexuals seeking sex reassignment is used as a model to investigate acute and chronic effects of testosterone and estradiol on serotonin reuptake transporter (SERT) binding in female- to-male and male-to-female transsexuals, suggesting that testosterone increases SERT expression on the cell surface.

Journal ArticleDOI
TL;DR: Observations performed in a subset of patients treated for male pattern hair loss indicate that persistent sexual side effects as well as anxious/depressive symptomatology have been reported even after discontinuation of finasteride treatment.

Journal ArticleDOI
TL;DR: Digit ratio is not related to fetal androgens or estrogens at late gestation, and there were no significant correlations between digit ratio and any androgen or estrogen concentrations considered individually.

Journal ArticleDOI
TL;DR: Sex differences in ERα‐IR and AR‐IR were observed in all hypothalamic regions analysed, although they most likely do not reflect the action of oestradiol because ArKO mice of both sexes showed expression levels very similar to wild‐type mice throughout perinatal development.
Abstract: Sex steroid hormones act on developing neural circuits regulating the hypothalamic-pituitary-gonadal axis and are involved in hormone-sensitive behaviours. These hormones act mainly via nuclear receptors, such as oestrogen receptor (ER)-α and androgen receptor (AR). By using immunohistochemistry, we analysed the expression level of ERα and AR throughout perinatal life [at embryonic (E) day 19 and postnatal (P) days 5, 15 and 25] and in adulthood in several hypothalamic nuclei controlling reproduction in both wild-type and aromatase knockout (ArKO) (i.e. which cannot convert testosterone into oestradiol) mice to determine whether there are sex differences in hypothalamic ERα and AR expression and, if so, whether these are established by the action of oestradiol. As early as E19, ERα immunoreactivity (-IR) was observed at same expression levels in both sexes in the anteroventral periventricular nucleus (AVPv), the medial preoptic area (MPOA), the bed nucleus of the stria terminalis (BnST), the ventrolateral part of the ventromedial hypothalamic nucleus and the arcuate nucleus (ARC). Sex differences (female > male) in ERα-IR were observed not only during the prepubertal period in the BnST (P5 to P25) and the MPOA (P15), but also in adulthood in these two brain regions. Sex differences in AR-IR (male > female) were observed at P5 in the AVPv and ARC, and at P25 in the MPOA and ARC, as well as in adulthood in all hypothalamic regions analysed. In adulthood, gonadectomy and hormonal treatment (oestradiol or dihydrotestosterone) also strongly modulated ERα-IR and AR, respectively. Taken together, sex differences in ERα-IR and AR-IR were observed in all hypothalamic regions analysed, although they most likely do not reflect the action of oestradiol because ArKO mice of both sexes showed expression levels very similar to wild-type mice throughout perinatal development.

Journal ArticleDOI
TL;DR: The results indicated that taurine can suppress testicular function deterioration by increasing antioxidant ability and inhibiting apoptosis.
Abstract: In males, the decline of androgen synthesis, spermatogenesis and sexual function are the main phenotypes of aging, which may be attributed to testicular dysfunction. Taurine can act as an antioxidant, a testosterone secretion stimulator, a sperm membrane stabilizer and motility factor, and an anti-apoptotic agent. Recent observational studies suggested that taurine may play an important role in spermatogenesis, but to date whether taurine has anti-aging effects on testes remains unknown. We found that in aged rats testicular SDH and G6PDH activities, marker enzymes of testes, serum testosterone, testicular 3β-HSD and 17β-HSD mRNA expression levels were significantly increased by taurine treatment. Taurine administration also markedly raised the sperm count, viability and motility, decreased the sperm abnormality. Our data suggested that taurine can postpone testicular function deterioration in aged rats. Importantly, we observed obvious elevation of testicular antioxidant enzymes (SOD, GSH, GSH-Px) activities, and remarkable reduction of ROS and MDA by taurine administration, indicating taurine can decrease testicular oxidative stress and lipid peroxidation in aged rats. Finally, we found taurine effectively reduced testicular DNA fragmentation, increased testicular Bcl-2 protein expression, and decreased cytochrome c, Bax, Fas, FasL and caspase-3 expression, suggesting taurine can prohibit aged testicular apoptosis by mitochondrial dependent and independent signal pathway. In summary, our results indicated that taurine can suppress testicular function deterioration by increasing antioxidant ability and inhibiting apoptosis.

Journal ArticleDOI
TL;DR: Fasting increased corticosterone and decreased testosterone in circulation, indicating that the testes can integrate and respond to cues of stress directly, and local inhibition of testosterone synthesis may allow for rapid and reversible changes in physiology and behavior when conditions are inappropriate for breeding.

Journal ArticleDOI
TL;DR: AGD in adult rats exhibits a degree of plasticity, which may be mediated by modulation of local androgen/estrogen action, and is concluded as a lifelong easily accessible biomarker of overall androgen action during the MPW.
Abstract: Androgen action during the fetal masculinization programming window (MPW) determines the maximum potential for growth of androgen-dependent organs (eg, seminal vesicles, prostate, penis, and perineum) and is reflected in anogenital distance (AGD). As such, determining AGD in postnatal life has potential as a lifelong easily accessible biomarker of overall androgen action during the MPW. However, whether the perineum remains androgen responsive in adulthood and thus responds plastically to perturbed androgen drive remains unexplored. To determine this, we treated adult male rats with either the antiandrogen flutamide or the estrogen diethylstilbestrol (DES) for 5 weeks, followed by a 4-week washout period of no treatment. We determined AGD and its correlate anogenital index (AGI) (AGD relative to body weight) at weekly intervals across this period and compared these with normal adult rats (male and female), castrated male rats, and appropriate vehicle controls. These data showed that, in addition to reducing circulating testosterone and seminal vesicle weight, castration significantly reduced AGD (by ∼17%), demonstrating that there is a degree of plasticity in AGD in adulthood. Flutamide treatment increased circulating testosterone yet also reduced seminal vesicle weight due to local antagonism of androgen receptor. Despite this suppression, surprisingly, flutamide treatment had no effect on AGD at any time point. In contrast, although DES treatment suppressed circulating testosterone and reduced seminal vesicle weight, it also induced a significant reduction in AGD (by ∼11%), which returned to normal 1 week after cessation of DES treatment. We conclude that AGD in adult rats exhibits a degree of plasticity, which may be mediated by modulation of local androgen/estrogen action. The implications of these findings regarding the use of AGD as a lifelong clinical biomarker of fetal androgen action are discussed.

Journal ArticleDOI
TL;DR: The data underscore the likely important role of androgens in the dynamics of folliculogenesis and challenges the idea that androgens induce high AMH levels, which is gaining more interest nowadays as an important particular PCOS feature.