Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 379
Filtrar
1.
Endocr Pract ; 30(4): 348-355, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38244859

RESUMO

OBJECTIVE: We determined (1) if 11-oxygenated androgens better identify polycystic ovary syndrome (PCOS) diagnosis in women with obesity compared to total or free testosterone (T) and free androgen index; (2) how biochemical hyperandrogenism and metabolic factors cluster in a cohort of women with infertility and obesity. METHODS: Women with obesity and PCOS comprised the study group (N = 132). Ovulatory women with obesity and idiopathic, tubal or male factor infertility were the control group (N = 83). Steroid hormones were measured by means of liquid chromatography tandem mass spectrometry. Receiver operating characteristic curves and principal component analysis were used. RESULTS: Women with obesity and PCOS had higher 11-ketotestosterone (11 KT) (1.22 nmol/L [0.84; 1.65] vs 1.05 [0.78; 1.35], P = .04) compared to controls, but not 11ß-hydroxyandrostenedione 4.30 [2.87; 5.92] vs 4.06 [3.22; 5.73], P = .44). 11-ketotestosterone (area under the curve: 0.59) did not better discriminate PCOS in women with obesity compared to: total T (0.84), free T (0.91), and free androgen index (0.85). We identified 4 principal components (PCs) in the PCOS group (72.1% explained variance): (1) insulin resistance status; (2) blood pressure; (3) obesity; (4) androgen status and 4 PCs in the control group (68.7% explained variance) with variables representing metabolism being dispersed in component 2, 3, and 4. CONCLUSIONS: Eleven-oxygenated androgens do not aid in the diagnosis of PCOS in women with obesity. Insulin resistance is the strongest PC in the PCOS group. There is no major dominant characteristic that defines obese non-PCOS women.


Assuntos
Hiperandrogenismo , Infertilidade , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Masculino , Humanos , Síndrome do Ovário Policístico/complicações , Hiperandrogenismo/diagnóstico , Hiperandrogenismo/metabolismo , Androgênios , Testosterona , Obesidade/complicações , Obesidade/metabolismo , Análise por Conglomerados
2.
Endocrinol Diabetes Metab ; 7(1): e443, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37872876

RESUMO

INTRODUCTION: Polycystic ovary syndrome (PCOS) is a common endocrine pathology in women. In addition to infertility, women with PCOS have metabolic dysregulation which predisposes them to Type 2 diabetes, cardiovascular disease and non-alcoholic fatty liver disease. Moreover, women with PCOS have changes in their gut microbial community that may be indicative of dysbiosis. While hyperandrogenism is associated with both the development of metabolic dysfunction and gut dysbiosis in females, the mechanisms involved are not well understood. METHODS: We used dihydrotestosterone (DHT) and ovariectomy (OVX) mouse models coupled with metabolic assessments and 16S rRNA gene sequencing to explore the contributions of hyperandrogenism and oestrogen deficiency to the development of insulin resistance and gut microbial dysbiosis in pubertal female mice. RESULTS: We demonstrated that, while DHT treatment or OVX alone were insufficient to induce insulin resistance during the pubertal-to-adult transition, combining OVX with DHT resulted in insulin resistance similar to that observed in letrozole-treated mice with elevated testosterone and decreased oestrogen levels. In addition, our results showed that OVX and DHT in combination resulted in a distinct shift in the gut microbiome compared to DHT or OVX alone, suggesting that the substantial metabolic dysregulation occurring in the OVX + DHT model was accompanied by unique changes in the abundances of gut bacteria including S24-7, Rikenellaceae and Mucispirillum schaedleri. CONCLUSIONS: While hyperandrogenism plays an important role in the development of metabolic dysregulation in female mice, our results indicate that investigation into additional factors influencing insulin resistance and the gut microbiome during the pubertal-to-adult transition could provide additional insight into the pathophysiology of PCOS.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Humanos , Adulto , Feminino , Camundongos , Animais , Hiperandrogenismo/complicações , Hiperandrogenismo/metabolismo , Disbiose/complicações , Disbiose/metabolismo , Diabetes Mellitus Tipo 2/complicações , RNA Ribossômico 16S , Síndrome do Ovário Policístico/complicações , Estrogênios
3.
Am J Pathol ; 193(12): 1916-1935, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37689383

RESUMO

Pregnancy-related problems have been linked to impairments in maternal uterine spiral artery (SpA) remodeling. The mechanisms underlying this association are still unclear. It is also unclear whether hyperandrogenism and insulin resistance, the two common manifestations of polycystic ovary syndrome, affect uterine SpA remodeling. We verified previous work in which exposure to 5-dihydrotestosterone (DHT) and insulin (INS) in rats during pregnancy resulted in hyperandrogenism, insulin intolerance, and higher fetal mortality. Exposure to DHT and INS dysregulated the expression of angiogenesis-related genes in the uterus and placenta and also decreased expression of endothelial nitric oxide synthase and matrix metallopeptidases 2 and 9, increased fibrotic collagen deposits in the uterus, and reduced expression of marker genes for SpA-associated trophoblast giant cells. These changes were related to a greater proportion of unremodeled uterine SpAs and a smaller proportion of highly remodeled arteries in DHT + INS-exposed rats. Placentas from DHT + INS-exposed rats exhibited decreased basal and labyrinth zone regions, reduced maternal blood spaces, diminished labyrinth vascularity, and an imbalance in the abundance of vascular and smooth muscle proteins. Furthermore, placentas from DHT + INS-exposed rats showed expression of placental insufficiency markers and a significant increase in cell senescence-associated protein levels. Altogether, this work demonstrates that increased pregnancy complications in polycystic ovary syndrome may be mediated by problems with uterine SpA remodeling, placental functionality, and placental senescence.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Humanos , Ratos , Gravidez , Feminino , Animais , Placenta/metabolismo , Síndrome do Ovário Policístico/metabolismo , Hiperandrogenismo/metabolismo , Útero/metabolismo , Artérias , Di-Hidrotestosterona/metabolismo , Insulina , Artéria Uterina/metabolismo
4.
Drug Res (Stuttg) ; 73(8): 441-447, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37591259

RESUMO

Poly cystic ovary syndrome (PCOS) is considered as one of the common hormonal disorders affecting 6-20% of women in their reproductive age with characteristic features include anovulatory infertility, hyperandrogenism, cystic follicles and insulin resistance. The gene CYP play an important role in pathophysiology of hyperandrogenism associated with PCOS. An elevated androgens are reported in PCOS condition due to overexpression of the enzyme CYP450 17 α: . As well as diminished levels of aromatase (CYP450 19) were observed in several hyperandrogenic PCOS patients. The powdered leafy material of Cinnamomum malabatrum was subjected to Soxhlet extraction. The plant extract was subjected to Gas chromatography-MS analysis (GC-MS), and the chromatogram obtained revealed the presence of active chemical constituents like 1(10),9(11)-B-Homolanistadiene for the first time and other potential compounds. Hypothesis has raised to interpret the efficiency of phytoconstituents of Cinnamomum malabatrum on these enzyme targets and which may be a novel drug candidate for the treatment and maintenance of hyperandrogenism associated with PCOS. Thus, the results obtained from the in-silico study of Cinnamomum malabatrum leaf extract using computational approaches indicate that the phytoconstituents have good affinities for the selected two key targets. ADME and PASS studies has been performed for active phytoconstituents homolanistadiene, ß-sitosterol, cycloartenol and a pyrazole derivative, and results revealed the Lipinski drug-likeness and pharmacological potential. In conclusion, this work throws a new insight into the possibility of the active phytoconstituents on binding the two active CYP45017 α and CYP45019 aromatase enzymes which facilitates development of novel compounds for hyperandrogenism associated with PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Hiperandrogenismo/tratamento farmacológico , Hiperandrogenismo/complicações , Hiperandrogenismo/metabolismo , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Aromatase , Cromatografia Gasosa-Espectrometria de Massas
5.
Int J Mol Sci ; 24(13)2023 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-37445796

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by hyperandrogenemia of ovarian thecal cell origin, resulting in anovulation/oligo-ovulation and infertility. Our previous studies established that ovarian theca cells isolated and propagated from ovaries of normal ovulatory women and women with PCOS have distinctive molecular and cellular signatures that underlie the increased androgen biosynthesis in PCOS. To evaluate differences between gene expression in single-cells from passaged cultures of theca cells from ovaries of normal ovulatory women and women with PCOS, we performed single-cell RNA sequencing (scRNA-seq). Results from these studies revealed differentially expressed pathways and genes involved in the acquisition of cholesterol, the precursor of steroid hormones, and steroidogenesis. Bulk RNA-seq and microarray studies confirmed the theca cell differential gene expression profiles. The expression profiles appear to be directed largely by increased levels or activity of the transcription factors SREBF1, which regulates genes involved in cholesterol acquisition (LDLR, LIPA, NPC1, CYP11A1, FDX1, and FDXR), and GATA6, which regulates expression of genes encoding steroidogenic enzymes (CYP17A1) in concert with other differentially expressed transcription factors (SP1, NR5A2). This study provides insights into the molecular mechanisms underlying the hyperandrogenemia associated with PCOS and highlights potential targets for molecular diagnosis and therapeutic intervention.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/metabolismo , Análise da Expressão Gênica de Célula Única , Hiperandrogenismo/complicações , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Fatores de Transcrição/genética
6.
Horm Behav ; 153: 105392, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37295324

RESUMO

Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine and metabolic abnormalities such as obesity and insulin resistance. PCOS is also associated with psychiatric disorders and cognitive impairment. The animal model of PCOS was induced by treating rats with 5α-dihydrotestosterone (5α-DHT) and additionally modified to induce adiposity by litter size reduction (LSR). Spatial learning and memory were assessed using the Barnes Maze test, and striatal markers of synaptic plasticity were analyzed. Striatal insulin signaling was estimated by the levels of insulin receptor substrate 1 (IRS1), its inhibitory phosphorylation at Ser307, and glycogen synthase kinase-3α/ß (GSK3α/ß) activity. Both LSR and DHT treatment significantly decreased striatal protein levels of IRS1, followed by increased GSK3α/ß activity in small litters. Results of the behavioral study showed that LSR had a negative effect on learning rate and memory retention, whereas DHT treatment did not induce impairment in memory formation. While protein levels of synaptophysin, GAP43, and postsynaptic density protein 95 (PSD-95) were not altered by the treatments, DHT treatment induced an increase in phosphorylation of PSD-95 at Ser295 in both normal and small litters. This study revealed that LSR and DHT treatment suppressed insulin signaling by downregulating IRS1 in the striatum. However, DHT treatment did not have an adverse effect on learning and memory, probably due to compensatory elevation in pPSD-95-Ser295, which had a positive effect on synaptic strength. This implies that hyperandrogenemia in this setting does not represent a threat to spatial learning and memory, opposite to the effect of overnutrition-related adiposity.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Ratos , Animais , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/metabolismo , Hiperandrogenismo/complicações , Hiperandrogenismo/metabolismo , Aprendizagem Espacial , Resistência à Insulina/fisiologia , Insulina/metabolismo , Di-Hidrotestosterona/farmacologia , Obesidade/complicações , Modelos Animais de Doenças
8.
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
9.
Placenta ; 137: 70-77, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37087951

RESUMO

INTRODUCTION: Trophoblast mitochondria play important roles in placental energy metabolism, physiology and pathophysiology. Hyperandrogenism has been associated with mitochondrial abnormalities in pregnancy disorders such as pre-eclampsia, gestational diabetes, and intrauterine growth restriction, but the direct impacts of androgen exposure on placental mitochondrial function are unknown. Given the inherent limitations of studying the human placenta during pregnancy, trophoblast cell lines are routinely used to model placental biology in vitro. The aim of this study was to characterize mitochondrial respiratory function in four commonly used trophoblast cell lines to provide a basis for selecting one well-suited to investigating the impact of androgens on trophoblast mitochondrial function. METHODS: Androgen receptor expression, mitochondrial respiration (JO2) and reactive oxygen species (ROS) release rates were evaluated in three human trophoblast cell lines (ACH-3P, BeWo and Swan-71) and one immortalized ovine trophoblast line (iOTR) under basal and substrate-stimulated conditions using high-resolution fluorespirometry. RESULTS: ACH-3P cells exhibited the greatest mitochondrial respiratory capacity and coupling efficiency of the four trophoblast lines tested, along with robust expression of androgen receptor protein that was found to co-localize with mitochondria by immunoblot and immunofluorescence. Acute testosterone administration (10 nM) tended to decrease ACH-3P mitochondrial JO2 and increase ROS release, while chronic (7 days) testosterone exposure increased expression of mitochondrial proteins, JO2, and ROS release. DISCUSSION: These studies establish ACH-3P as a suitable cell line for investigating trophoblast mitochondrial function, and provide foundational evidence supporting links between hyperandrogenism and placental mitochondrial ROS production with potential relevance to several common pregnancy disorders.


Assuntos
Hiperandrogenismo , Trofoblastos , Gravidez , Feminino , Animais , Ovinos , Humanos , Trofoblastos/metabolismo , Placenta/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Androgênicos/metabolismo , Testosterona/farmacologia , Testosterona/metabolismo , Hiperandrogenismo/metabolismo , Mitocôndrias/metabolismo
10.
Iran J Med Sci ; 48(2): 187-197, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36895462

RESUMO

Background: Polycystic ovary syndrome (PCOS) is the most common reproductive dysfunction in premenopausal women. PCOS is associated with oxidative stress (OS), which is the main risk factor for renal diseases. This study aimed to investigate the mechanisms responsible for renal injury in a hyperandrogenemic female rat model. Methods: This study was conducted from December 2019 to September 2021 at Shiraz Nephro-Urology Research Centre, Shiraz University of Medical Sciences (Shiraz, Iran). Thirty female Sprague-Dawley rats were randomly divided into three groups (n=10), namely control, sham, and dehydroepiandrosterone (DHEA). Plasma total testosterone, plasma creatinine (Cr), and blood urea nitrogen (BUN) levels were measured. In addition, total oxidant status (TOS), total antioxidant capacity (TAC), oxidative stress index (OSI), and histopathological changes in the ovaries and kidneys were determined. Data were analyzed using the GraphPad Prism software, and P<0.05 was considered statistically significant. Results: Plasma total testosterone levels increased by nine-fold in DHEA-treated rats compared to controls (P=0.0001). Administration of DHEA increased Cr and BUN levels and caused severe renal tubular cell injury. In addition, plasma and tissue (kidney and ovary) TAC levels decreased significantly, but TOS levels and OSI values were significantly increased (P=0.019). Significant damage to both glomerular and tubular parts of the kidney and ovarian follicular structure was observed in the DHEA group. Conclusion: Hyperandrogenemia caused systemic abnormalities through OS-related mechanisms and damaged renal and ovarian tissues. DHEA treatment in rat models is recommended to study the mechanisms that mediate PCOS-associated renal injury.


Assuntos
Hiperandrogenismo , Nefropatias , Síndrome do Ovário Policístico , Humanos , Ratos , Feminino , Animais , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Ratos Sprague-Dawley , Hiperandrogenismo/complicações , Hiperandrogenismo/metabolismo , Hiperandrogenismo/patologia , Estresse Oxidativo , Rim , Antioxidantes/metabolismo , Nefropatias/patologia , Testosterona/metabolismo , Desidroepiandrosterona/metabolismo
11.
Mymensingh Med J ; 32(1): 3-9, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36594292

RESUMO

Polycystic ovary syndrome (PCOS) is a polygenic and multifactorial condition, regarded as the most common endocrine abnormality of women in reproductive period. It is commonly assumed that insulin resistance, hyperandrogenism and obesity significantly influence the pathophysiological process of PCOS. This study was designed to estimate hormonal parameters in different phenotypes of PCOS. The cross sectional descriptive type of observational study was carried out at Mymensingh Medical College Hospital, Mymensingh, Bangladesh from January 2018 to June 2019. Data were collected from purposively selected 107 patients with PCOS by interview, clinical examination and laboratory investigations using a pretested case record form. Data were analyzed by computer software, SPSS-version 22.0. Hormonal parameters in different phenotypes of PCOS were compared with ANOVA test. Phenotype A was found in highest number (59.8%) followed by phenotype B (14.9%), phenotype D (14.0%) and phenotype C (11.2%). Biochemical hyperandrogenism was observed highest in phenotype A (57.8%) followed by phenotype B (36.4%) and phenotype C (6.1%). Biochemical or clinical hyperandrogenism was not observed among patients of phenotype D. Altered LH:FSH ratio was high in phenotype A (14.1%) and Phenotype B (2.8%). Increased serum prolactin level was found highest in phenotype A (10.3%) and increased serum TSH was found highest in phenotype D (4.7%). Statistically significant difference was observed among levels of serum testosterone of different phenotypes (p<0.001). Hormonal derangements among different phenotypes reflect the severity of reproductive dysfunction and metabolic aberrations. Screening for metabolic risks of diverse phenotypes is important to detect and prevent long term health consequences of PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Estudos Transversais , Hiperandrogenismo/metabolismo , Fenótipo
12.
Curr Nutr Rep ; 12(1): 191-202, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36719550

RESUMO

PURPOSE OF REVIEW: Polycystic ovary syndrome (PCOS), which is common in women of reproductive age worldwide, is a syndrome that reduces the lifelong quality of life and poses a significant risk for various diseases. PCOS is a combination of symptoms of hyperandrogenism, oligo-anovulation, and polycystic ovarian morphology (PCOM). In PCOS, which is characterized by chronic low-grade inflammation, some inflammatory cytokines are increased. This review aimed to explain possible mechanisms of inflammation in PCOS and the effects of Mediterranean diet components on reducing this inflammation. RECENT FINDINGS: Although the exact mechanisms of inflammation in PCOS are not yet fully known, it is stated that it is mediated by obesity, insulin resistance, and high androgen concentration. This inflammatory state negatively impacts the risk of future health problems and the quality of life of PCOS. Therefore, strategies to reduce inflammation are thought to be important. Dietary adjustments have important effects in reducing this inflammation and preventing disease. At this point, the Mediterranean diet, which has been proven to have a protective effect against many diseases, draws attention. Among the components of the Mediterranean diet, especially omega-3, antioxidants and dietary fiber may contribute to the reduction of inflammation through different mechanisms. PCOS is characterized by chronic low-grade inflammation, which increases women's risk of health problems, both now and in the future. Reducing inflammation is therefore extremely important, and it can be achieved with adherence to the Mediterranean diet. Inflammation pathways and the effect of the components of the Mediterranean diet in PCOS. AGE, advanced glycation end products; NF-κB, nuclear factor kappa-B. Obesity, insulin resistance, and hyperandrogenism may cause inflammation in PCOS through different mechanisms. Antioxidants, omega-3, and dietary fiber, which are the main components of the Mediterranean diet, may be effective in reducing this inflammation in PCOS. (Created with BioRender.com).


Assuntos
Dieta Mediterrânea , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/metabolismo , Hiperandrogenismo/metabolismo , Qualidade de Vida , Inflamação , Obesidade , Fibras na Dieta
13.
Life Sci ; 313: 121224, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36435224

RESUMO

AIMS: Polycystic ovary syndrome (PCOS) is a common endocrine disorder in the women of childbearing age. It is characterized by hyperandrogenism and abnormal follicular growth and ovulation. The polyol pathway is a glucose metabolism bypass pathway initiated by aldose reductase (ADR). Androgen induces the expression of ADR in the male reproductive tract, which has a general physiological significance for male reproductive function. Here we investigate whether hyperandrogenemia in PCOS leads to increased flux of the polyol pathway in ovarian tissue, which in turn affects follicular maturation and ovulation through oxidative stress. MAIN METHODS: We used clinical epidemiological methods to collect serum and granulosa cells from clinical subjects for a clinical case-control study. At the same time, cell biology and molecular biology techniques were used to conduct animal and cell experiments to further explore the mechanism of hyperandrogen-induced ovarian polyol pathway hyperactivity and damage to ovarian function. KEY FINDINGS: Here, we find that hyperandrogenism of PCOS can induce the expression of ovarian aldose reductase, which leads to the increase of the polyol pathway flux, and affects ovarian function through excessive oxidative stress. SIGNIFICANCE: Our research has enriched the pathological mechanism of PCOS and may provide a new clue for the clinical treatment of PCOS.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Humanos , Animais , Feminino , Masculino , Síndrome do Ovário Policístico/metabolismo , Hiperandrogenismo/metabolismo , Aldeído Redutase/metabolismo , Estudos de Casos e Controles , Estresse Oxidativo
14.
BMC Mol Cell Biol ; 23(1): 47, 2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36368943

RESUMO

BACKGROUND: Although hormonal and metabolic dysfunction have been recognized as a possible cause of polycystic ovarian syndrome (PCOS), the associations between hyperandrogenism and aryl hydrocarbon receptor (Ahr) signaling pathway remains controversial. The current study aimed to investigate the effect of hyperandrogenism on oocyte developmental competency via regarding Ahr signaling downstream pathway in granulosa cells. MATERIALS AND METHODS: Granulosa cells were collected from 45 PCOS patients under assisted reproductive technique (ART). Gene expression of Ahr downstream pathway was evaluated based on Reverse Transcription Q-PCR assay. Moreover the correlation was investigated between gene expression and hyperandrogenism, and oocyte developmental competency in PCOS. RESULTS: From the 45 PCOS patients, 26 (64.44%) had a high level of follicular fluid testosterone (FFT). Based on the FFT level, two groups of PCOS: HFT (high level of FFT) and non-HFT, were shown significant differences in oocyte and embryo quality, and fertilization and cleavage rates. Moreover, the mean relative expressions of Ahr and Arnt genes were significantly higher in HFT -PCOS group (p < 0.01 and p < 0.01) respectively. Also, the significant positive correlations were obtained for Ahr, Arnt, Cyp1A1, and Cyp1B1 with incidence of clinical hyperandrogenism and FFT level. Besides, our results showed that Ahr, Cyp1A1, and Cyp1B1 gene expression was correlated significantly with fertilization rate. CONCLUSION: The present study suggested that hyperandrogenism could impair oocyte developmental competency via affecting Ahr signaling downstream pathway.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Líquido Folicular/metabolismo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Hiperandrogenismo/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Testosterona , Oócitos/metabolismo
15.
Mol Metab ; 65: 101583, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36096453

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrine disorder, defined by reproductive and endocrine abnormalities, with metabolic dysregulation including obesity, insulin resistance and hepatic steatosis. Recently, it was found that skeletal muscle insulin sensitivity could be improved in obese, post-menopausal, pre-diabetic women through treatment with nicotinamide mononucleotide (NMN), a precursor to the prominent redox cofactor nicotinamide adenine dinucleotide (NAD+). Given that PCOS patients have a similar endocrine profile to these patients, we hypothesised that declining NAD levels in muscle might play a role in the pathogenesis of the metabolic syndrome associated with PCOS, and that this could be normalized through NMN treatment. Here, we tested the impact of NMN treatment on the metabolic syndrome of the dihydrotestosterone (DHT) induced mouse model of PCOS. We observed lower NAD levels in the muscle of PCOS mice, which was normalized by NMN treatment. PCOS mice were hyperinsulinaemic, resulting in increased adiposity and hepatic lipid deposition. Strikingly, NMN treatment completely normalized these aspects of metabolic dysfunction. We propose that addressing the decline in skeletal muscle NAD levels associated with PCOS can normalize insulin sensitivity, preventing compensatory hyperinsulinaemia, which drives obesity and hepatic lipid deposition, though we cannot discount an impact of NMN on other tissues to mediate these effects. These findings support further investigation into NMN treatment as a new therapy for normalizing the aberrant metabolic features of PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome Metabólica , Síndrome do Ovário Policístico , Animais , Di-Hidrotestosterona/metabolismo , Feminino , Humanos , Hiperandrogenismo/metabolismo , Resistência à Insulina/fisiologia , Lipídeos , Síndrome Metabólica/metabolismo , Camundongos , Músculo Esquelético/metabolismo , NAD/metabolismo , Mononucleotídeo de Nicotinamida/metabolismo , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo
16.
Am J Physiol Endocrinol Metab ; 323(2): E145-E158, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35658542

RESUMO

Polycystic ovary syndrome (PCOS) is a common, multifactorial disorder characterized by endocrine, reproductive, and metabolic dysfunction. As the etiology of PCOS is unknown, there is no cure and symptom-oriented treatments are suboptimal. Hyperandrogenism is a key diagnostic trait, and evidence suggests that androgen receptor (AR)-mediated actions are critical to PCOS pathogenesis. However, the key AR target sites involved remain to be fully defined. Adipocyte and muscle dysfunction are proposed as important sites involved in the manifestation of PCOS traits. We investigated the role of AR signaling in white adipose tissue (WAT), brown adipose tissue (BAT), and skeletal muscle in the development of PCOS in a hyperandrogenic PCOS mouse model. As expected, dihydrotestosterone (DHT) exposure induced key reproductive and metabolic PCOS traits in wild-type (WT) females. Transplantation of AR-insensitive (AR-/-) WAT or BAT from AR knockout females (ARKO) into DHT-treated WT mice ameliorated some metabolic PCOS features, including increased body weight, adiposity, and adipocyte hypertrophy, but not reproductive PCOS traits. In contrast, DHT-treated ARKO female mice transplanted with AR-responsive (AR+/+) WAT or BAT continued to resist developing PCOS traits. DHT-treated skeletal muscle-specific AR knockout females (SkMARKO) displayed a comparable phenotype with that of DHT-treated WT females, with full development of PCOS traits. Taken together, these findings infer that both WAT and BAT, but less likely skeletal muscle, are key sites of AR-mediated actions involved in the experimental pathogenesis of metabolic PCOS traits. These data further support targeting adipocyte AR-driven pathways in future research aimed at developing novel therapeutic interventions for PCOS.NEW & NOTEWORTHY Hyperandrogenism is a key feature in the pathogenesis of polycystic ovary syndrome (PCOS); however, the tissue sites of androgen receptor (AR) signaling are unclear. In this study, AR signaling in white and brown adipose tissue, but less likely in skeletal muscle, was found to be involved in the development of metabolic PCOS traits, highlighting the importance of androgen actions in adipose tissue and obesity in the manifestation of metabolic disturbances.


Assuntos
Tecido Adiposo Marrom , Tecido Adiposo , Androgênios , Hiperandrogenismo , Síndrome do Ovário Policístico , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/metabolismo , Androgênios/farmacologia , Animais , Di-Hidrotestosterona/farmacologia , Modelos Animais de Doenças , Feminino , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Fenótipo , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/genética
17.
Reprod Biol Endocrinol ; 20(1): 88, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35701786

RESUMO

BACKGROUND: According to current definitions of Polycystic Ovary Syndrome (PCOS), hyperandrogenism is considered as a key element in the pathogenesis of this common endocrinopathy. However, until now, studies about ovarian androgen profile in women are very rare. Our aim was then to characterise the expression profile of the androgens in follicular fluid of 30 PCOS patients, and compare it to those of 47 Control women and 29 women with only polycystic ovary morphology on ultrasounds (ECHO group). METHODS: A retrospective, single-centre cohort study was performed. The intrafollicular concentrations of the key androgens were assessed and correlated with the intrafollicular levels of some adipokines of interest. Androgens were quantified by mass spectrophotometry combined with ultra-high-performance liquid chromatography, while adipokine concentrations were measured by ELISA assays. RESULTS: In PCOS patients, the intrafollicular concentrations of the androgens synthesised by ovarian theca cells, i.e., 17OH-pregnenolone, dehydroepiandrosterone, Δ4-androstenedione and testosterone, were significantly higher than those of the androgens of adrenal origin, and positively correlated with the main PCOS clinical and biological features, as well as with the adipokines mostly expressed in the follicular fluid of PCOS patients, i.e. resistin, omentin, chemerin and apelin. Conversely, Control women showed the highest levels of 17OH-progesterone, deoxycorticosterone and 11-deoxycortisol. Confirming these results, apelin levels were negatively associated with pregnenolone and deoxycorticosterone concentrations, while visfatin levels, which were higher in the Control group, negatively correlated with the Δ4-androstenedione and testosterone ones. CONCLUSIONS: PCOS is characterised by a selective increase in the intrafollicular levels of the androgens synthesised by theca cells, strengthening the hypothesis that ovarian hyperandrogenism plays a central role in its pathogenesis. Further, the significant correlation between the intrafollicular concentrations of the androgens and most of the adipokines of interest, including apelin, chemerin, resistin and omentin, confirms the existence of a close relationship between these two hormonal systems, which appear deeply involved in ovarian physiology and PCOS physiopathology.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Adipocinas , Androgênios/metabolismo , Androstenodiona/metabolismo , Apelina , Estudos de Coortes , Desoxicorticosterona , Feminino , Líquido Folicular/metabolismo , Humanos , Hiperandrogenismo/metabolismo , Síndrome do Ovário Policístico/metabolismo , Pregnenolona , Resistina , Estudos Retrospectivos , Testosterona
18.
Front Endocrinol (Lausanne) ; 13: 868572, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757434

RESUMO

Obesity, altered glucose homeostasis, hyperinsulinism, and reproductive dysfunction develops in female humans and mammals with hyperandrogenism. We previously reported that low dose dihydrotestosterone (DHT) administration results in metabolic and reproductive dysfunction in the absence of obesity in female mice, and conditional knock-out of the androgen receptor (Ar) in the liver (LivARKO) protects female mice from DHT-induced glucose intolerance and hyperinsulinemia. Since altered metabolic function will regulate reproduction, and liver plays a pivotal role in the reversible regulation of reproductive function, we sought to determine the reproductive phenotype of LivARKO mice under normal and hyperandrogenemic conditions. Using Cre/Lox technology, we deleted the Ar in the liver, and we observed LivARKO female mice have normal puberty timing, cyclicity and reproductive function. After DHT treatment, like control mice, LivARKO experience altered estrous cycling, reduced numbers of corpus lutea, and infertility. Liver Ar is not involved in hyperandrogenemia-induced reproductive dysfunction. The reproductive dysfunction in the DHT-treated LivARKO lean females with normal glucose homeostasis indicates that androgen-induced reproductive dysfunction is independent from metabolic dysfunction.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Receptores Androgênicos , Reprodução , Animais , Di-Hidrotestosterona/farmacologia , Feminino , Glucose/metabolismo , Humanos , Hiperandrogenismo/induzido quimicamente , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Hiperandrogenismo/fisiopatologia , Fígado/metabolismo , Camundongos , Obesidade/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/deficiência , Receptores Androgênicos/metabolismo , Reprodução/fisiologia
19.
Reprod Biol Endocrinol ; 20(1): 91, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729637

RESUMO

BACKGROUND: Kisspeptin released from Kiss-1 neurons in the hypothalamus plays an essential role in the control of the hypothalamic-pituitary-gonadal axis by regulating the release of gonadotropin-releasing hormone (GnRH). In this study, we examined how androgen supplementation affects the characteristics of Kiss-1 neurons. METHODS: We used a Kiss-1-expressing mHypoA-55 cell model that originated from the arcuate nucleus (ARC) of the mouse hypothalamus. These cells are KNDy neurons that co-express neurokinin B (NKB) and dynorphin A (DynA). We stimulated these cells with androgens and examined them. We also examined the ARC region of the hypothalamus in ovary-intact female rats after supplementation with androgens. RESULTS: Stimulation of mHypoA-55 cells with 100 nM testosterone significantly increased Kiss-1 gene expression by 3.20 ± 0.44-fold; testosterone also increased kisspeptin protein expression. The expression of Tac3, the gene encoding NKB, was also increased by 2.69 ± 0.64-fold following stimulation of mHypoA-55 cells with 100 nM testosterone. DynA gene expression in these cells was unchanged by testosterone stimulation, but it was significantly reduced at the protein level. Dihydrotestosterone (DHT) had a similar effect to testosterone in mHypoA-55 cells; kisspeptin and NKB protein expression was significantly increased by DHT, whereas it significantly reduced DynA expression. In ovary-intact female rats, DTH administration significantly increased the gene expression of Kiss-1 and Tac3, but not DynA, in the arcuate nucleus. Exogenous NKB and DynA stimulation failed to modulate Kiss-1 gene expression in mHypoA-55 cells. Unlike androgen stimulation, prolactin stimulation did not modulate kisspeptin, NKB, or DynA protein expression in these cells. CONCLUSIONS: Our observations imply that hyperandrogenemia affects KNDy neurons and changes their neuronal characteristics by increasing kisspeptin and NKB levels and decreasing DynA levels. These changes might cause dysfunction of the hypothalamic-pituitary-gonadal axis.


Assuntos
Dinorfinas , Hiperandrogenismo , Androgênios/metabolismo , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hiperandrogenismo/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Neurocinina B/genética , Neurocinina B/metabolismo , Neurocinina B/farmacologia , Neurônios/metabolismo , Ratos , Taquicininas , Testosterona/metabolismo , Testosterona/farmacologia
20.
Am J Reprod Immunol ; 88(2): e13580, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35598286

RESUMO

Polycystic ovarian syndrome (PCOS) is a complicated neuro-endocrinal, reproductive, and metabolic condition. It encompasses patterns such as hyperandrogenism, recurrent cysts triggered by steroidogenic functional aberrations in the ovaries, overweight, chronic inflammation, and more. The underlying cause of this heterogeneous illness is obscure, although it is suspected to be driven by a blend of environmental and hereditary factors. In recent years, the connection between the microbiome and PCOS has been acknowledged and is thought to be involved in the genesis of the syndrome's emergence. Microbiota vary in different pathological features of PCOS, and fundamental pathways linked to their involvement in the commencement of diverse clinical presentations in PCOS open up a new avenue for its management. Prebiotic, probiotic, synbiotic, and fecal-microbiota-transplant, by promoting eubiosis and nullifying the effect caused by the altered microbial profile in PCOS women, can aid in management of diverse phenotypes associated with the syndrome. These microbiota-mediated treatments improve PCOS women's metabolic, inflammatory, and hormonal profiles. However, more studies are needed to elucidate the mechanisms that drive this positive effect.


Assuntos
Hiperandrogenismo , Microbiota , Síndrome do Ovário Policístico , Feminino , Humanos , Hiperandrogenismo/metabolismo , Síndrome do Ovário Policístico/metabolismo , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...