Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Geroscience ; 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38776045

RESUMO

17α-Estradiol (17αE2), a less-feminising enantiomer of 17ß-estradiol, has been shown to prolong lifespan and improve metabolic health in a sex-specific manner in male, but not in female mice. Recent studies have demonstrated the pivotal role of estrogen receptor α (ERα) in mediating the effects of 17αE2 on metabolic health. However, the specific tissues and/or neuronal signalling pathways that 17αE2 acts through remain to be elucidated. ERα expression in glutamatergic and GABAergic neurons (principal excitatory and inhibitory neurons respectively) in the hypothalamus is essential for estradiol signalling. Therefore, we hypothesised that knocking out ERα from one of these neuronal populations would attenuate the established beneficial metabolic effects of 17αE2 in male mice exposed to a high fat diet. To test this hypothesis we used two established brain specific ERα KO models, targeting either glutamatergic or GABAergic neurons (Vglut2/Vgat-ERαKO). We show that both of these ERα KO models exhibit a strong reduction in ERα expression in the arcuate nucleus of the hypothalamus, a control centre for metabolic regulation. Deletion of ERα from GABAergic neurons significantly diminished the effect of 17αE2 on body weight relative to controls, although these animals still show metabolic benefits with 17αE2 treatment. The response to 17αE2 was unaffected by ERα deletion in glutamatergic neurons. Our results support a benefit of 17αE2 treatment in protection against metabolic dysfunction, but these effects do not depend on exclusive ERα expression in glutamatergic and GABAergic neurons and persist when ERα expression is strongly reduced in the arcuate nucleus of the hypothalamus.

2.
J Neurosci ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744532

RESUMO

Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, that mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with POMC neurons, which regulate satiety and are affected by obesity, we examined their crosstalk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.Significance Statement Obesity presents a significant health concern, with multiple comorbidities, including impaired reproduction. However, mechanisms are not clear, and studies are confounded by the chronic nature of this condition that leads to synaptic changes and alterations in neuron responsiveness to stimuli. Here, we demonstrate that the interaction between feeding circuitry and reproductive circuitry is altered by chronic obesity. The reason may be that chronically higher activity of POMC neurons in response to higher leptin in obesity, downregulates αMSH receptors on target neurons, including kisspeptin. This may lead to the suppression of kisspeptin neurons, and their inability to regulate pulsatile secretion of GnRH, which then lowers LH pulse frequency, leading to lower LH in the circulation, lower testosterone, and lower sperm count.

4.
J Neuroendocrinol ; 36(3): e13370, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38344844

RESUMO

Excess levels of circulating androgens during prenatal or peripubertal development are an important cause of polycystic ovary syndrome (PCOS), with the brain being a key target. Approximately half of the women diagnosed with PCOS also experience metabolic syndrome; common features including obesity, insulin resistance and hyperinsulinemia. Although a large amount of clinical and preclinical evidence has confirmed this relationship between androgens and the reproductive and metabolic features of PCOS, the mechanisms by which androgens cause this dysregulation are unknown. Neuron-specific androgen receptor knockout alleviates some PCOS-like features in a peripubertal dihydrotestosterone (DHT) mouse model, but the specific neuronal populations mediating these effects are undefined. A candidate population is the agouti-related peptide (AgRP)-expressing neurons, which are important for both reproductive and metabolic function. We used a well-characterised peripubertal androgenized mouse model and Cre-loxP transgenics to investigate whether deleting androgen receptors specifically from AgRP neurons can alleviate the induced reproductive and metabolic dysregulation. Androgen receptors were co-expressed in 66% of AgRP neurons in control mice, but only in <2% of AgRP neurons in knockout mice. The number of AgRP neurons was not altered by the treatments. Only 20% of androgen receptor knockout mice showed rescue of DHT-induced androgen-induced anovulation and acyclicity. Furthermore, androgen receptor knockout did not rescue metabolic dysfunction (body weight, adiposity or glucose and insulin tolerance). While we cannot rule out developmental compensation in our model, these results suggest peripubertal androgen excess does not markedly influence Agrp expression and does not dysregulate reproductive and metabolic function through direct actions of androgens onto AgRP neurons.


Assuntos
Androgênios , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Camundongos , Gravidez , Proteína Relacionada com Agouti/metabolismo , Androgênios/metabolismo , Di-Hidrotestosterona/farmacologia , Camundongos Knockout , Neurônios/metabolismo , Obesidade/metabolismo , Peptídeos/farmacologia , Receptores Androgênicos/metabolismo , Virilismo/metabolismo
5.
J Endocr Soc ; 7(10): bvad113, 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37693686

RESUMO

Immunohistochemical visualization of progesterone receptor (PR)-expressing cells in the brain is a powerful technique to investigate the role of progesterone in the neuroendocrine regulation of fertility. A major obstacle to the immunohistochemical visualization of progesterone-sensitive cells in the rodent brain has been the discontinuation of the commercially produced A0098 rabbit polyclonal PR antibody by DAKO. To address the unavailability of this widely used PR antibody, we optimized and evaluated 4 alternative commercial PR antibodies and found that each lacked the specificity and/or sensitivity to immunohistochemically label PR-expressing cells in paraformaldehyde-fixed female mouse brain sections. As a result, we developed and validated a new custom RC269 PR antibody, directed against the same 533-547 amino acid sequence of the human PR as the discontinued A0098 DAKO PR antibody. Immunohistochemical application of the RC269 PR antibody on paraformaldehyde-fixed mouse brain sections resulted in nuclear PR labeling that was highly distinguishable from background, specific to its antigen, highly regulated by estradiol, matched the known distribution of PR protein expression in the female mouse hypothalamus, and nearly identical to that of the discontinued A0098 DAKO PR antibody. In summary, the RC269 PR antibody is a specific and sensitive antibody to immunohistochemically visualize PR-expressing cells in the mouse brain.

6.
JMIR Form Res ; 7: e44810, 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37624626

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) is a heterogeneous condition that affects 4% to 21% of people with ovaries. Inaccessibility or dissatisfaction with clinical treatment for PCOS has led to some individuals with the condition discussing their experiences in specialized web-based forums. OBJECTIVE: This study explores the feasibility of using such web-based forums for clinical research purposes by gathering and analyzing laboratory test results posted in an active PCOS forum, specifically the PCOS subreddit hosted on Reddit. METHODS: We gathered around 45,000 posts from the PCOS subreddit. A random subset of 5000 posts was manually read, and the presence of laboratory test results was labeled. These labeled posts were used to train a machine learning model to identify which of the remaining posts contained laboratory results. The laboratory results were extracted manually from the identified posts. These self-reported laboratory test results were compared with values in the published literature to assess whether the results were concordant with researcher-published values for PCOS cohorts. A total of 10 papers were chosen to represent published PCOS literature, with selection criteria including the Rotterdam diagnostic criteria for PCOS, a publication date within the last 20 years, and at least 50 participants with PCOS. RESULTS: Overall, the general trends observed in the laboratory test results from the PCOS web-based forum were consistent with clinically reported PCOS. A number of results, such as follicle stimulating hormone, fasting insulin, and anti-Mullerian hormone, were concordant with published values for patients with PCOS. The high consistency of these results among the literature and when compared to the subreddit suggests that follicle stimulating hormone, fasting insulin, and anti-Mullerian hormone are more consistent across PCOS phenotypes than other test results. Some results, such as testosterone, sex hormone-binding globulin, and homeostasis model assessment-estimated insulin resistance index, were between those of PCOS literature values and normal values, as defined by clinical testing limits. Interestingly, other results, including dehydroepiandrosterone sulfate, luteinizing hormone, and fasting glucose, appeared to be slightly more dysregulated than those reported in the literature. CONCLUSIONS: The differences between the forum-posted results and those published in the literature may be due to the selection process in clinical studies and the possibility that the forum disproportionally describes PCOS phenotypes that are less likely to be alleviated with medical intervention. However, the degree of concordance in most laboratory test values implied that the PCOS web-based forum participants were representative of research-identified PCOS cohorts. This validation of the PCOS subreddit grants the possibility for more research into the contents of the subreddit and the idea of undertaking similar research using the contents of other medical internet forums.

7.
J Neuroendocrinol ; 35(6): e13302, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280378

RESUMO

Polycystic ovary syndrome (PCOS) is a female endocrine disorder that is associated with prenatal exposure to excess androgens. In prenatally androgenized (PNA) mice that model PCOS, GABAergic neural transmission to and innervation of GnRH neurons is increased. Evidence suggests that elevated GABAergic innervation originates in the arcuate nucleus (ARC). We hypothesized that GABA-GnRH circuit abnormalities are a direct consequence of PNA, resulting from DHT binding to androgen receptor (AR) in the prenatal brain. However, whether prenatal ARC neurons express AR at the time of PNA treatment is presently unknown. We used RNAScope in situ hybridization to localize AR mRNA (Ar)-expressing cells in healthy gestational day (GD) 17.5 female mouse brains and to assess coexpression levels in specific neuronal phenotypes. Our study revealed that less than 10% of ARC GABA cells expressed Ar. In contrast, we found that ARC kisspeptin neurons, critical regulators of GnRH neurons, were highly colocalized with Ar. Approximately 75% of ARC Kiss1-expressing cells also expressed Ar at GD17.5, suggesting that ARC kisspeptin neurons are potential targets of PNA. Investigating other neuronal populations in the ARC we found that ~50% of pro-opiomelanocortin (Pomc) cells, 22% of tyrosine hydroxylase (Th) cells, 8% of agouti-related protein (Agrp) cells and 8% of somatostatin (Sst) cells express Ar. Lastly, RNAscope in coronal sections showed Ar expression in the medial preoptic area (mPOA), and the ventral part of the lateral septum (vLS). These Ar-expressing regions were highly GABAergic, and 22% of GABA cells in the mPOA and 25% of GABA cells in the vLS also expressed Ar. Our findings identify specific neuronal phenotypes in the ARC, mPOA, and vLS that are androgen sensitive in late gestation. PNA-induced functional changes in these neurons may be related to the development of impaired central mechanisms associated with PCOS-like features.


Assuntos
Androgênios , Síndrome do Ovário Policístico , Humanos , Camundongos , Feminino , Gravidez , Animais , Androgênios/metabolismo , Receptores Androgênicos/metabolismo , Kisspeptinas/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios GABAérgicos/fisiologia , Encéfalo/metabolismo , Virilismo/metabolismo
8.
Reproduction ; 166(3): R51-R61, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37345882

RESUMO

In brief: Neuroendocrine dysfunction and transgenerational susceptibility associated with polycystic ovary syndrome (PCOS) suggest that programmed changes within the brain contribute to adult development of the syndrome. This review discusses a potentially important role for microglia in mediating prenatal androgen-programmed changes in the female brain that contribute to PCOS-like features. Abstract: Several lines of evidence support a role for the brain in both the development and maintenance of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility worldwide. Persistently elevated luteinizing hormone secretion and impaired gonadal steroid hormone feedback in PCOS patients suggest impairments within the neuronal networks that regulate the reproductive axis. Evidence from preclinical models has linked androgen excess during prenatal life with altered structure and function of the developing female brain that might underpin syndrome development in adulthood. Studies investigating the mechanisms by which excess androgens program changes in the female brain have highlighted an important role for microglia. This review discusses how these non-neuronal cells shape the developing female brain in response to excess androgens and focuses on how microglia may be involved in the development of the neuroendocrine dysfunctions associated with PCOS.

9.
Endocrinology ; 164(6)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37191144

RESUMO

Polycystic ovarian syndrome (PCOS) is the leading cause of anovulatory infertility and is a heterogenous condition associated with a range of reproductive and metabolic impairments. While its etiology remains unclear, hyperandrogenism and impaired steroid negative feedback have been identified as key factors underpinning the development of PCOS-like features both clinically and in animal models. We tested the hypothesis that androgen signaling in kisspeptin-expressing neurons, which are key drivers of the neuroendocrine reproductive axis, is critically involved in PCOS pathogenesis. To this end, we used a previously validated letrozole (LET)-induced hyperandrogenic mouse model of PCOS in conjunction with Cre-lox technology to generate female mice exhibiting kisspeptin-specific deletion of androgen receptor (KARKO mice) to test whether LET-treated KARKO females are protected from the development of reproductive and metabolic PCOS-like features. LET-treated mice exhibited hyperandrogenism, and KARKO mice exhibited a significant reduction in the coexpression of kisspeptin and androgen receptor mRNA compared to controls. In support of our hypothesis, LET-treated KARKO mice exhibited improved estrous cyclicity, ovarian morphology, and insulin sensitivity in comparison to LET-treated control females. However, KARKO mice were not fully protected from the effects of LET-induced hyperandrogenism and still exhibited reduced corpora lutea numbers and increased body weight gain. These data indicate that increased androgen signaling in kisspeptin-expressing neurons plays a critical role in PCOS pathogenesis but highlight that other mechanisms are also involved.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Animais , Feminino , Camundongos , Androgênios/metabolismo , Modelos Animais de Doenças , Hiperandrogenismo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Letrozol , Neurônios/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo
10.
Mol Hum Reprod ; 29(6)2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37171897

RESUMO

Currently, our understanding of hormonal regulation within the female reproductive system is largely based on our knowledge of estrogen and progesterone signalling. However, while the important functions of androgens in male physiology are well known, it is also recognized that androgens play critical roles in the female reproductive system. Further, androgen signalling is altered in a variety of gynaecological conditions, including endometriosis and polycystic ovary syndrome, indicative of regulatory roles in endometrial and ovarian function. Co-regulatory mechanisms exist between different androgens, estrogens, and progesterone, resulting in a complex network of steroid hormone interactions. Evidence from animal knockout studies, in vitro experiments, and human data indicate that androgen receptor expression is cell-specific and menstrual cycle stage-dependent, with important regulatory roles in the menstrual cycle, endometrial biology, and follicular development in the ovaries. This review will discuss the expression and co-regulatory interactions of androgen receptors, highlighting the complexity of the androgen signalling pathway in the endometrium and ovaries, and the synthesis of androgens from additional alternative pathways previously disregarded as male-specific. Moreover, it will illustrate the challenges faced when studying androgens in female biology, and the need for a more in-depth, integrative view of androgen metabolism and signalling in the female reproductive system.


Assuntos
Androgênios , Ovário , Animais , Masculino , Feminino , Humanos , Ovário/metabolismo , Progesterona/metabolismo , Endométrio/metabolismo , Estrogênios , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo
11.
Front Endocrinol (Lausanne) ; 14: 1116482, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875467

RESUMO

Introduction: Polycystic ovary syndrome (PCOS) is the most common infertility disorder worldwide, typically characterised by high circulating androgen levels, oligo- or anovulation, and polycystic ovarian morphology. Sexual dysfunction, including decreased sexual desire and increased sexual dissatisfaction, is also reported by women with PCOS. The origins of these sexual difficulties remain largely unidentified. To investigate potential biological origins of sexual dysfunction in PCOS patients, we asked whether the well-characterized, prenatally androgenized (PNA) mouse model of PCOS exhibits modified sex behaviours and whether central brain circuits associated with female sex behaviour are differentially regulated. As a male equivalent of PCOS is reported in the brothers of women with PCOS, we also investigated the impact of maternal androgen excess on the sex behaviour of male siblings. Methods: Adult male and female offspring of dams exposed to dihydrotestosterone (PNAM/PNAF) or an oil vehicle (VEH) from gestational days 16 to 18 were tested for a suite of sex-specific behaviours. Results: PNAM showed a reduction in their mounting capabilities, however, most of PNAM where able to reach ejaculation by the end of the test similar to the VEH control males. In contrast, PNAF exhibited a significant impairment in the female-typical sexual behaviour, lordosis. Interestingly, while neuronal activation was largely similar between PNAF and VEH females, impaired lordosis behaviour in PNAF was unexpectedly associated with decreased neuronal activation in the dorsomedial hypothalamic nucleus (DMH). Conclusion: Taken together, these data link prenatal androgen exposure that drives a PCOS-like phenotype with altered sexual behaviours in both sexes.


Assuntos
Lordose , Síndrome do Ovário Policístico , Feminino , Masculino , Gravidez , Humanos , Animais , Camundongos , Androgênios , Irmãos , Virilismo
12.
Curr Opin Pharmacol ; 68: 102345, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36621270

RESUMO

Polycystic ovary syndrome is a prevalent endocrinopathy involving androgen excess, and anovulatory infertility. The disorder is also associated with many comorbidities such as obesity and hyperinsulinemia, and an increased risk of cardiovascular complications. Reproductive, endocrine, and metabolic symptoms are highly variable, with heterogenous phenotypes adding complexity to clinical management of symptoms. This review highlights recent findings regarding emerging therapies for treating polycystic ovary syndrome, including i) pharmacological agents to target androgen excess, ii) modulation of kisspeptin signalling to target central neuroendocrine dysregulation, and iii) novel insulin sensitisers to combat peripheral metabolic dysfunction.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/metabolismo , Androgênios , Comorbidade , Obesidade/tratamento farmacológico , Obesidade/complicações
13.
Front Endocrinol (Lausanne) ; 13: 1017468, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36457554

RESUMO

Polycystic ovary syndrome (PCOS) affects up to 20% of women but remains poorly understood. It is a heterogeneous condition with many potential comorbidities. This review offers an overview of the dysregulation of the reproductive and metabolic systems associated with PCOS. Review of the literature informed the development of a comprehensive summarizing 'wiring' diagram of PCOS-related features. This review provides a justification for each diagram aspect from the relevant academic literature, and explores the interactions between the hypothalamus, ovarian follicles, adipose tissue, reproductive hormones and other organ systems. The diagram will provide an efficient and useful tool for those researching and treating PCOS to understand the current state of knowledge on the complexity and variability of PCOS.


Assuntos
Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/etiologia , Folículo Ovariano , Reprodução , Tecido Adiposo , Hormônios
14.
Curr Opin Pharmacol ; 67: 102321, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36427399

RESUMO

Reproductive function is tightly regulated by both environmental and physiological factors. The adipose-derived hormone leptin has been identified as one such critical factor that relays information about peripheral energy availability to the centrally-governed HPG axis to ensure there is sufficient energy availability to support the high energy demands of mammalian reproduction. In the absence of adequate central leptin signaling, reproductive function is suppressed. While leptin levels are predominantly regulated by adiposity, circulating leptin levels are also under the modulatory influence of other factors, such as stress system activation, circadian rhythmicity, and immune activation and the inflammatory response. Furthermore, changes in leptin sensitivity can affect the degree to which leptin exerts its influence on the neuroendocrine reproductive axis. This review will discuss the different mechanisms by which leptin serves to integrate and relay information about metabolic, psychological, environmental and immune conditions to the central neuronal network that governs reproductive function.


Assuntos
Leptina , Reprodução , Animais , Humanos , Leptina/metabolismo , Reprodução/fisiologia , Obesidade , Transdução de Sinais , Mamíferos/metabolismo
15.
J Neuroendocrinol ; 34(12): e13206, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36416198

RESUMO

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy to affect women of reproductive-age world-wide. Hyperandrogenism is both a hallmark feature of PCOS, and is hypothesised to be an underlying mechanism driving the development of the condition in utero. With circulating hormones known to profoundly influence maternal responses in females, we aimed to determine whether maternal behaviour is altered in a well-described prenatally androgenised (PNA) mouse model of PCOS. Mouse dams were administered with dihydrotestosterone or vehicle on days 16, 17 and 18 of pregnancy. Maternal responses were assessed in both the dihydrotestosterone-injected dams following parturition and in their adult female PNA offspring. Exposure of dams to excess androgens during late pregnancy had no detrimental effects on pregnancy outcomes, including gestation length, pup survival and gestational weight gain, or on subsequent maternal behaviour following parturition. By contrast, PNA virgin females, modelling PCOS, exhibited enhanced maternal behaviour when tested in an anxiogenic novel cage environment, with females rapidly retrieving pups and nesting with them. In comparison, most control virgin females failed to complete this retrieval task in the anxiogenic environment. Assessment of progesterone receptor and oestrogen receptor α immunoreactivity in the brains of virgin PNA and control females revealed increased numbers of oestrogen receptor α positive cells in the brains of PNA females in regions well known to be important for maternal behaviour. This suggests that increased oestrogenic signalling in the neural circuit that underlies maternal behaviour may be a possible mechanism by which maternal behaviour is enhanced in PNA female mice.


Assuntos
Di-Hidrotestosterona , Comportamento Materno , Síndrome do Ovário Policístico , Animais , Feminino , Camundongos , Gravidez , Androgênios/farmacologia , Di-Hidrotestosterona/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Síndrome do Ovário Policístico/induzido quimicamente , Reprodução , Virilismo/metabolismo , Comportamento Materno/efeitos dos fármacos , Comportamento Materno/fisiologia
16.
Front Endocrinol (Lausanne) ; 13: 951344, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992143

RESUMO

Polycystic ovary syndrome (PCOS) is associated with elevated androgen and luteinizing hormone (LH) secretion and with oligo/anovulation. Evidence indicates that elevated androgens impair sex steroid hormone feedback regulation of pulsatile LH secretion. Hyperandrogenemia in PCOS may also disrupt the preovulatory LH surge. The mechanisms through which this might occur, however, are not fully understood. Kisspeptin (KISS1) neurons of the rostral periventricular area of the third ventricle (RP3V) convey hormonal cues to gonadotropin-releasing hormone (GnRH) neurons. In rodents, the preovulatory surge is triggered by these hormonal cues and coincident timing signals from the central circadian clock in the suprachiasmatic nucleus (SCN). Timing signals are relayed to GnRH neurons, in part, via projections from SCN arginine-vasopressin (AVP) neurons to RP3VKISS1 neurons. Because rodent SCN cells express androgen receptors (AR), we hypothesized that these circuits are impaired by elevated androgens in a mouse model of PCOS. In prenatally androgen-treated (PNA) female mice, SCN Ar expression was significantly increased compared to that found in prenatally vehicle-treated mice. A similar trend was seen in the number of Avp-positive SCN cells expressing Ar. In the RP3V, the number of kisspeptin neurons was preserved. Anterograde tract-tracing, however, revealed reduced SCNAVP neuron projections to the RP3V and a significantly lower proportion of RP3VKISS1 neurons with close appositions from SCNAVP fibers. Functional assessments showed, on the other hand, that RP3VKISS1 neuron responses to AVP were maintained in PNA mice. These findings indicate that PNA changes some of the neural circuits that regulate the preovulatory surge. These impairments might contribute to ovulatory dysfunction in PNA mice modeling PCOS.


Assuntos
Kisspeptinas , Síndrome do Ovário Policístico , Núcleo Supraquiasmático , Androgênios/metabolismo , Androgênios/farmacologia , Animais , Arginina , Arginina Vasopressina/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Camundongos , Neurônios/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Gravidez , Núcleo Supraquiasmático/metabolismo , Vasopressinas/metabolismo
17.
J Neuroendocrinol ; 34(5): e13115, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35502534

RESUMO

The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.


Assuntos
Hormônio Liberador de Gonadotropina , Neuroanatomia , Animais , Hormônio Liberador de Gonadotropina/metabolismo , Mamíferos , Neurônios/metabolismo , Prosencéfalo , Reprodução
18.
Compr Physiol ; 12(2): 3347-3369, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35578968

RESUMO

Polycystic ovary syndrome (PCOS) is a major endocrine disorder strongly associated with androgen excess and frequently leading to female infertility. Although classically considered an ovarian disease, altered neuroendocrine control of gonadotropin-releasing hormone (GnRH) neurons in the brain and abnormal gonadotropin secretion may underpin PCOS presentation. Defective regulation of GnRH pulse generation in PCOS promotes high luteinizing hormone (LH) pulsatile secretion, which in turn overstimulates ovarian androgen production. Early and emerging evidence from preclinical models suggests that maternal androgen excess programs abnormalities in developing neuroendocrine circuits that are associated with PCOS pathology, and that these abnormalities are sustained by postpubertal elevation of endogenous androgen levels. This article will discuss experimental evidence, from the clinic and in preclinical animal models, that has significantly contributed to our understanding of how androgen excess influences the assembly and maintenance of neuroendocrine impairments in the female brain. Abnormal central gamma-aminobutyric acid (GABA) signaling has been identified in both patients and preclinical models as a possible link between androgen excess and elevated GnRH/LH secretion. Enhanced GABAergic innervation and drive to GnRH neurons is suspected to contribute to the pathogenesis and early manifestation of neuroendocrine derangement in PCOS. Accordingly, this article also provides an overview of GABA regulation of GnRH neuron function from prenatal development to adulthood to discuss possible avenues for future discovery research and therapeutic interventions. © 2022 American Physiological Society. Compr Physiol 12:3347-3369, 2022.


Assuntos
Síndrome do Ovário Policístico , Adulto , Androgênios , Animais , Feminino , Hormônio Liberador de Gonadotropina , Humanos , Sistemas Neurossecretores , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/patologia , Gravidez , Ácido gama-Aminobutírico
19.
J Neuroendocrinol ; 34(4): e13110, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35267218

RESUMO

Polycystic ovary syndrome (PCOS) is associated with androgen excess and, frequently, hyperactive pulsatile luteinizing hormone (LH) secretion. Although the origins of PCOS are unclear, evidence from pre-clinical models implicates androgen signalling in the brain in the development of PCOS pathophysiology. Chronic exposure of female mice to dihydrotestosterone (DHT) from 3 weeks of age drives both reproductive and metabolic impairments that are ameliorated by selective androgen receptor (AR) loss from the brain. This suggests centrally driven mechanisms in hyperandrogen-mediated PCOS-like pathophysiology that remain to be defined. Acute prenatal DHT exposure can also model the hyperandrogenism of PCOS, and this is accompanied by increased LH pulse frequency and increased GABAergic innervation of gonadotrophin-releasing hormone (GnRH) neurons. We aimed to determine the impact of chronic exposure of female mice to DHT, which models the hyperandrogenism of PCOS, on pulsatile LH secretion and putative GABAergic input to GnRH neurons. To do this, GnRH-green fluorescent protein (GFP) female mice received either DHT or blank capsules for 90 days from postnatal day 21 (n = 6 or 7 per group). Serial tail-tip blood sampling was used to measure LH dynamics and perfusion-fixed brains were collected and immunolabelled for vesicular GABA transporter (VGAT) to assess putative GABAergic terminals associated with GFP-labelled GnRH neurons. As expected, chronic DHT resulted in acyclicity and significantly increased body weight. However, no differences in LH pulse frequency or the density of VGAT appositions to GnRH neurons were identified between ovary-intact DHT-treated females and controls. Chronic DHT exposure significantly increased the number of AR expressing cells in the hypothalamus, whereas oestrogen receptor α-expressing neuron number was unchanged. Therefore, although chronic DHT exposure from 3 weeks of age increases AR expressing neurons in the brain, the GnRH neuronal network changes and hyperactive LH secretion associated with prenatal androgen excess are not evident. These findings suggest that unique central mechanisms are involved in the reproductive impairments driven by exposure to androgen excess at different developmental stages.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Efeitos Tardios da Exposição Pré-Natal , Androgênios/metabolismo , Animais , Di-Hidrotestosterona , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hiperandrogenismo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo
20.
J Neuroendocrinol ; 34(5): e13093, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35083794

RESUMO

Given the critical central role of gonadotropin-releasing hormone (GnRH) neurons in fertility, it is not surprising that the GnRH neural network is implicated in the pathology of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility. Although many symptoms of PCOS relate most proximately to ovarian dysfunction, the central reproductive neuroendocrine system ultimately drives ovarian function through its regulation of anterior pituitary gonadotropin release. The typical cyclical changes in frequency of GnRH release are often absent in women with PCOS, resulting in a persistent high-frequency drive promoting gonadotropin changes (i.e., relatively high luteinizing hormone and relatively low follicle-stimulating hormone concentrations) that contribute to ovarian hyperandrogenemia and ovulatory dysfunction. However, the specific mechanisms underpinning GnRH neuron dysfunction in PCOS remain unclear. Here, we summarize several preclinical and clinical studies that explore the causes of aberrant GnRH secretion in PCOS and the role of disordered GnRH secretion in PCOS pathophysiology.


Assuntos
Hormônio Liberador de Gonadotropina , Síndrome do Ovário Policístico , Feminino , Hormônio Foliculoestimulante , Gonadotropinas , Humanos , Hormônio Luteinizante , Neurônios
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...