Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.839
Filtrar
Más filtros

Intervalo de año de publicación
1.
Biol Cell ; 116(7): e2300069, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38679788

RESUMEN

Polycystic ovary syndrome or PCOS is an endocrine disorder in women of reproductive age. It is a diversified multi factorial disorder and diagnosis is very complicated because of its overlapping symptoms some of which are irregular menstrual cycle, acne in face, excess level of androgen (AE), insulin resistance, obesity, cardiovascular disease, mood disorder and type 2 diabetes (T2DM). PCOS may be caused by hormonal imbalance, genetic and epigenetic vulnerability, hypothalamic and ovarian troubles. PCOS is essentially hyperandrogenimia with oligo-anovulation. This review explains the abnormal regulation of autophagy related genes and proteins in different cells at various stages which leads to the genesis of PCOS. During nutrient starvation cells face stress condition, which it tries to overcome by activating its macroautophagy mechanism and by degrading the cytoplasmic material. This provides energy to the cell facilitating its survival. Downregulation of autophagy related genes in endometria has been observed in PCOS women. PCOS can be managed by maintaining proper lifestyle and medical treatment. Healthy meals and regular exercise can prevent the excessive weight and also reduce the PCOS complications. Medicines such as metformin, clomiphene, and the oral contraceptive pill can also balance the hormonal level. The imbalance in regulation of autophagy genes has been discussed with correlation to PCOS. The different management strategies for PCOS have also been summarized.


Asunto(s)
Autofagia , Síndrome del Ovario Poliquístico , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Humanos , Femenino , Animales
2.
Cell Mol Life Sci ; 81(1): 101, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38409361

RESUMEN

Abnormal autophagy is one of the vital features in polycystic ovary syndrome (PCOS). However, the underlying molecular mechanisms remain unelucidated. In this study, we aimed to investigate whether Block of Proliferation 1 (BOP1) is involved in the onset of autophagy activation of granulosa cells in PCOS. Firstly, we found that BOP1 expression was significantly down-regulated in the ovaries of PCOS mice, which was associated with the development of PCOS. Next, local injection of lentiviral vectors in the ovary for the overexpression of BOP1 significantly alleviated the phenotypes of elevated androgens, disturbed estrous cycle, and abnormal follicular development in PCOS mice. Subsequently, we found that knockdown of BOP1 activated autophagy of granulosa cells in the in vitro experiments, whereas overexpression of BOP1 inhibited autophagy in both in vivo and in vitro models. Mechanistically, BOP1 knockdown triggered the nucleolus stress response, which caused RPL11 to be released from the nucleolus into the nucleoplasm and inhibited the E3 ubiquitination ligase of MDM2, thereby enhancing the stability of p53. Subsequently, P53 inhibited mTOR, thereby activating autophagy in granulosa cells. In addition, the mRNA level of BOP1 was negatively correlated with antral follicle count (AFC), body-mass index (BMI), serum androgen levels, and anti-Mullerian hormone (AMH) in patients with PCOS. In summary, our study demonstrates that BOP1 downregulation inhibits mTOR phosphorylation through activation of the p53-dependent nucleolus stress response, which subsequently contributes to aberrant autophagy in granulosa cells, revealing that BOP1 may be a key target for probing the mechanisms of PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratones , Células de la Granulosa/metabolismo , Folículo Ovárico/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(30): e2203503119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867816

RESUMEN

Women with polycystic ovary syndrome (PCOS) frequently experience decreased sexual arousal, desire, and sexual satisfaction. While the hypothalamus is known to regulate sexual behavior, the specific neuronal pathways affected in patients with PCOS are not known. To dissect the underlying neural circuitry, we capitalized on a robust preclinical animal model that reliably recapitulates all cardinal PCOS features. We discovered that female mice prenatally treated with anti-Müllerian hormone (PAMH) display impaired sexual behavior and sexual partner preference over the reproductive age. Blunted female sexual behavior was associated with increased sexual rejection and independent of sex steroid hormone status. Structurally, sexual dysfunction was associated with a substantial loss of neuronal nitric oxide synthase (nNOS)-expressing neurons in the ventromedial nucleus of the hypothalamus (VMH) and other areas of hypothalamic nuclei involved in social behaviors. Using in vivo chemogenetic manipulation, we show that nNOSVMH neurons are required for the display of normal sexual behavior in female mice and that pharmacological replenishment of nitric oxide restores normal sexual performance in PAMH mice. Our data provide a framework to investigate facets of hypothalamic nNOS neuron biology with implications for sexual disturbances in PCOS.


Asunto(s)
Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico , Síndrome del Ovario Poliquístico , Conducta Sexual , Núcleo Hipotalámico Ventromedial , Animales , Hormona Antimülleriana/farmacología , Modelos Animales de Enfermedad , Femenino , Preferencia en el Apareamiento Animal , Ratones , Neuronas/efectos de los fármacos , Neuronas/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Síndrome del Ovario Poliquístico/enzimología , Síndrome del Ovario Poliquístico/fisiopatología , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/metabolismo
4.
J Cell Mol Med ; 28(8): e18258, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38546608

RESUMEN

Polycystic ovary syndrome (PCOS) is one of the modern intractable reproductive diseases. The female irregular menstruation, infertility, obesity, and so forth caused by PCOS have become a hot issue affecting family harmony and social development. The aetiology of PCOS is complex. In recent years, many scholars have found that its pathogenesis was related to the imbalance of gut microbiota. Gut microbiota can form two-way communication with the brain through the 'gut-brain axis' and affect the host's metabolism. Current research has confirmed that the gut microbiota can interfere with glucose and lipid metabolism, insulin sensitivity, hormone secretion and follicular development in women by altering intestinal mucosal permeability and secreting metabolites. In addition, the diversity and composition of gut microbiota of PCOS patients changed, which may affect the metabolic function of the gut microbiota and the ability to produce metabolites, and may also directly or indirectly affect the endocrine function. This study reviewed recent research advances about the role of gut microbiota in PCOS. In order to provide basis for prevention and treatment of PCOS based on gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , Humanos , Femenino , Eje Cerebro-Intestino , Inmunidad Innata , Transporte Biológico
5.
BMC Genomics ; 25(1): 208, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38408933

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a complex multifactorial disorder with a substantial genetic component. However, the clinical manifestations of PCOS are heterogeneous with notable differences between lean and obese women, implying a different pathophysiology manifesting in differential body mass index (BMI). We performed a meta-analysis of genome-wide association study (GWAS) data from six well-characterised cohorts, using a case-control study design stratified by BMI, aiming to identify genetic variants associated with lean and overweight/obese PCOS subtypes. RESULTS: The study comprised 254,588 women (5,937 cases and 248,651 controls) from individual studies performed in Australia, Estonia, Finland, the Netherlands and United States of America, and separated according to three BMI stratifications (lean, overweight and obese). Genome-wide association analyses were performed for each stratification within each cohort, with the data for each BMI group meta-analysed using METAL software. Almost half of the total study population (47%, n = 119,584) were of lean BMI (≤ 25 kg/m2). Two genome-wide significant loci were identified for lean PCOS, led by rs12000707 within DENND1A (P = 1.55 × 10-12) and rs2228260 within XBP1 (P = 3.68 × 10-8). One additional locus, LINC02905, was highlighted as significantly associated with lean PCOS through gene-based analyses (P = 1.76 × 10-6). There were no significant loci observed for the overweight or obese sub-strata when analysed separately, however, when these strata were combined, an association signal led by rs569675099 within DENND1A reached genome-wide significance (P = 3.22 × 10-9) and a gene-based association was identified with ERBB4 (P = 1.59 × 10-6). Nineteen of 28 signals identified in previous GWAS, were replicated with consistent allelic effect in the lean stratum. There were less replicated signals in the overweight and obese groups, and only 4 SNPs were replicated in each of the three BMI strata. CONCLUSIONS: Genetic variation at the XBP1, LINC02905 and ERBB4 loci were associated with PCOS within unique BMI strata, while DENND1A demonstrated associations across multiple strata, providing evidence of both distinct and shared genetic features between lean and overweight/obese PCOS-affected women. This study demonstrated that PCOS-affected women with contrasting body weight are not only phenotypically distinct but also show variation in genetic architecture; lean PCOS women typically display elevated gonadotrophin ratios, lower insulin resistance, higher androgen levels, including adrenal androgens, and more favourable lipid profiles. Overall, these findings add to the growing body of evidence supporting a genetic basis for PCOS as well as differences in genetic patterns relevant to PCOS BMI-subtype.


Asunto(s)
Estudio de Asociación del Genoma Completo , Síndrome del Ovario Poliquístico , Femenino , Humanos , Índice de Masa Corporal , Sobrepeso/genética , Estudios de Casos y Controles , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/complicaciones , Obesidad/genética
6.
Curr Issues Mol Biol ; 46(6): 5223-5241, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38920985

RESUMEN

Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder that affects women of reproductive age, representing the primary cause of anovulatory infertility. The nonapeptide oxytocin (OT) plays an important role in cognitive, emotional, and reproductive functions in human beings. Oxytocin receptors are expressed in several body parts, including the ovaries. Despite this, the possible role played by oxytocin in symptoms of PCOS is not clear. The present systematic review aimed at understanding the presence of possible oxytocin level alterations in PCOS, the connection between alterations of OT levels and the symptoms of PCOS, and the effect of oxytocin administration in PCOS. After a systematic search in the principal databases, eight studies, five human and three animal, were included. Four human studies and one animal study highlighted the role played by oxytocin in fertility issues related to PCOS. Three human and two animal studies investigated the role of body weight and OT levels. Studies that analyzed oxytocin basal levels in women agreed that PCOS is associated with a reduction in the serum level of oxytocin. Two human studies and one animal study agreed about lower levels of oxytocin, confirming a possible implication of the dysfunction of OT in the pathogenesis of PCOS.

7.
Curr Issues Mol Biol ; 46(4): 3379-3393, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38666942

RESUMEN

Polycystic ovary syndrome (PCOS) is often associated with metabolic abnormalities in the affected patients such as obesity or a dysregulated glucose metabolism/insulin resistance (IR). IR affects the serum levels of several circulating microRNAs; however, studies on the association between IR-related microRNAs and PCOS are scarce. Therefore, we quantified the serum levels of the IR-associated microRNAs miR-93, miR-148a, miR-216a, miR-224 and miR-320a via qPCR in a cohort of 358 infertility patients, of whom 136 were diagnosed with PCOS. In bivariate correlation analyses, the serum levels of miR-93 and miR-216a were inversely associated with dipeptidyl peptidase 4 serum concentrations, and the miR-320a serum levels were significantly downregulated in PCOS patients (p = 0.02, Mann-Whitney U test). Interestingly, in all patients who achieved pregnancy after Assisted Reproductive Technology (ART) cycles, the serum levels of the five IR-associated microRNAs were significantly elevated compared to those of non-pregnant patients. In cell culture experiments, we detected a significant upregulation of miR-320a expression following testosterone stimulation over 24 and 48 h in KGN and COV434 granulosa carcinoma cells. In conclusion, we demonstrated a significantly reduced serum level of the IR-associated miR-320a in our patient cohort. This result once again demonstrates the close relationship between metabolic disorders and the dysregulation of microRNA expression patterns in PCOS.

8.
Am J Epidemiol ; 193(10): 1399-1406, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-38751314

RESUMEN

Most previous studies found an elevated risk of endometrial cancer among women with polycystic ovary syndrome (PCOS). However, these had highly varying methods for ascertainment of PCOS diagnoses and limitations such as few exposed women and short follow-up. In this cohort study, we investigated the association between PCOS and endometrial cancer among women born in Denmark between January 1, 1940, and December 31, 1993 (n = 1 719 121). Data in this study, including PCOS and endometrial cancer diagnoses and covariates, were derived from nationwide registers. We used Cox proportional hazards regression models to calculate hazard ratios (HRs) and 95% CIs. A total of 7862 endometrial cancer cases were identified during 23.7 years of follow-up (IQR, 37.7-61.9). We found an increased risk of endometrial cancer among women with PCOS compared with women without PCOS (HR = 3.02; 95% CI, 2.03-4.49). The risk was increased for premenopausal women (HR = 5.82; 95% CI, 3.64-9.30), whereas no marked association was seen for postmenopausal women. However, for postmenopausal women, results were limited by few cases and young age at the end of follow-up. Mounting evidence of an increased risk for endometrial cancer among women with PCOS reinforces the need for prevention and early detection. This article is part of a Special Collection on Gynecological Cancers.


Asunto(s)
Neoplasias Endometriales , Síndrome del Ovario Poliquístico , Modelos de Riesgos Proporcionales , Humanos , Síndrome del Ovario Poliquístico/epidemiología , Síndrome del Ovario Poliquístico/complicaciones , Femenino , Neoplasias Endometriales/epidemiología , Dinamarca/epidemiología , Adulto , Persona de Mediana Edad , Factores de Riesgo , Estudios de Cohortes , Sistema de Registros , Anciano , Premenopausia , Posmenopausia
9.
Am J Epidemiol ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38960722

RESUMEN

OBJECTIVES: To assess correlates of diagnosed and probable polycystic ovary syndrome (PCOS) among parous women. METHODS: This study includes 557 women recruited from multi-specialty clinics in eastern Massachusetts. We categorized women as "diagnosed PCOS" based on medical records and self-reported clinician-diagnoses. Next, we constructed a category of "probable PCOS" for women without a diagnosis but with ≥2 of the following: ovulatory dysfunction (cycle length<21 or ≥35 days), hyperandrogenism (free testosterone>75th percentile), or elevated anti-Müllerian hormone (>75th percentile). We classified the remaining as "no PCOS," and compared characteristics across groups. RESULTS: 9.7% had diagnosed and 9.2% had probable PCOS. The frequency of irregular cycles was similar for diagnosed and probable PCOS. Free testosterone and AMH were higher for probable than diagnosed PCOS. Frequency of irregular cycles and both hormones were higher for the two PCOS groups vs. the no PCOS group. Obesity prevalence for diagnosed PCOS was twice that of probable PCOS (43.9% vs. 19.6%), yet the two groups had similar HbA1c and adiponectin. CONCLUSIONS: Women with probable PCOS are leaner but have comparable glycemic traits to those with a formal diagnosis, highlighting the importance of assessing biochemical profiles among women with irregular cycles, even in the absence of overweight/obesity.

10.
Funct Integr Genomics ; 24(5): 171, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39317806

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disease in women of childbearing age, making it imperative to explore more biomarkers for PCOS. Furthermore, previous studies have reported that cyclin dependent kinase inhibitor 1 C (CDKN1C) might be associated with PCOS progression. However, the molecular mechanism of CDKN1C involved in PCOS is poorly defined. METHODS: CDKN1C and Yin-Yang-1 (YY1) expression levels were determined using real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot assay. Cell viability, proliferation, cell cycle progression, and cell apoptosis were analyzed using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. Caspase 3 activity was examined using a commercial kit. Binding between YY1 and CDKN1C promoter was predicted by JASPAR and verified using Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays. RESULTS: CDKN1C and YY1 were highly expressed in PCOS granulosa cells (GCs). Furthermore, CDKN1C silencing could promote cell proliferation and cell cycle process and repress cell apoptosis in human ovarian granulosa cell line KGN cells. For mechanistic analysis, YY1 is directly bound to the promoter of CDKN1C and transcriptional-regulated CDKN1C expression. CONCLUSION: YY1-activated CDKN1C might block KGN cell proliferation and induce cell apoptosis, providing a possible therapeutic target for PCOS treatment.


Asunto(s)
Apoptosis , Proliferación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina , Células de la Granulosa , Síndrome del Ovario Poliquístico , Activación Transcripcional , Regulación hacia Arriba , Factor de Transcripción YY1 , Factor de Transcripción YY1/metabolismo , Factor de Transcripción YY1/genética , Humanos , Femenino , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Adulto , Regiones Promotoras Genéticas
11.
Biochem Biophys Res Commun ; 707: 149782, 2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38493745

RESUMEN

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder in women of reproductive age, which often leads to female infertility. Chronic inflammation is a significant factor in the development of PCOS. Our study aimed to explore the impact of mesencephalic astrocyte-derived neurotrophic factor (MANF), a scientifically validated anti-inflammatory factor, on 99 diagnosed PCOS patients. We also investigated its effects on PCOS mice induced with dehydroepiandrosterone (DHEA) and KGN cells induced with dihydrotestosterone (DHT). Our findings revealed a decrease in serum MANF levels in PCOS patients, which were negatively associated with serum tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) levels. The administration of recombinant human MANF (rhMANF) in PCOS mice demonstrated a decrease in pro-inflammatory cytokines and monocytes/macrophages in both peripheral blood and ovarian tissues. Furthermore, the inclusion of rhMANF notably ameliorated DHEA-induced ovarian dysfunction and fibrosis by negatively regulating the toll-like receptor 4 (TLR4)-nuclear factor kappa B (NF-κB)-NLR family, pyrin domain containing protein 3 (NLRP3) pathway. Additionally, in vitro experiments showed that the up-regulation of MANF offset DHT-induced inhibition of viability and apoptosis in KGN cells. Collectively, this study highlights the anti-inflammatory properties of MANF in PCOS and suggests its potential as a therapeutic approach for the management of PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico , Femenino , Humanos , Ratones , Animales , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/complicaciones , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 4 , Astrocitos/metabolismo , Antiinflamatorios/uso terapéutico , Factores de Crecimiento Nervioso , Deshidroepiandrosterona/farmacología , Deshidroepiandrosterona/uso terapéutico
12.
Artículo en Inglés | MEDLINE | ID: mdl-39167287

RESUMEN

PURPOSE: Although some reproductive and metabolic characteristics of polycystic ovary syndrome (PCOS) are known risk factors for breast cancer, the evidence regarding a potential association between PCOS and breast cancer is scarce. In this population-based cohort study including all 1,719,452 women born in Denmark between 1940 and 1993, we investigated the association between PCOS and breast cancer. METHODS: PCOS diagnoses, cancer diagnoses, covariates, migrations, and vital status were all obtained from national population and health registers. Hazard ratios (HR) and 95% confidence intervals (CI) for breast cancer overall and for histological subtypes separately were calculated based on adjusted cox proportional hazards models. RESULTS: During a median follow-up of 26 years, 63,078 women were diagnosed with breast cancer. We found an increased risk of breast cancer overall among women with PCOS compared with women without PCOS (HR: 1.21, 95% CI 1.02-1.44). In analyses stratified for menopausal status, the increased risk was restricted to postmenopausal women (HR: 1.63, 95% CI 1.23-2.15). The results for ductal and lobular histological subtypes analyses separately resembled those observed for breast cancer overall. CONCLUSION: This is the first study to report an increased risk of breast cancer among women with a history of PCOS. The increased risk was seemingly confined to postmenopausal women. Our results therefore contribute to an increased knowledge of the etiology of breast cancer, but our findings should be further confirmed in other large cohort studies with an appropriately long follow-up period.

13.
Biol Reprod ; 110(4): 782-797, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38224314

RESUMEN

Defining features of polycystic ovary syndrome (PCOS) include elevated expression of steroidogenic genes, theca cell androgen biosynthesis, and peripheral levels of androgens. In previous studies, we identified vascular cell adhesion molecule 1 (VCAM1) as a selective androgen target gene in specific NR2F2/SF1 (+/+) theca cells. By deleting NR2F2 and VCAM1 selectively in CYP17A1 theca cells in mice, we documented that NR2F2 and VCAM1 impact distinct and sometimes opposing theca cell functions that alter ovarian follicular development in vivo: including major changes in ovarian morphology, steroidogenesis, gene expression profiles, immunolocalization images (NR5A1, CYP11A1, NOTCH1, CYP17A1, INSL3, VCAM1, NR2F2) as well as granulosa cell functions. We propose that theca cells impact follicle integrity by regulating androgen production and action, as well as granulosa cell differentiation/luteinization in response to androgens and gonadotropins that may underlie PCOS.


Asunto(s)
Factor de Transcripción COUP II , Síndrome del Ovario Poliquístico , Células Tecales , Molécula 1 de Adhesión Celular Vascular , Animales , Femenino , Ratones , Andrógenos/metabolismo , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Células Tecales/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Blood Cells Mol Dis ; 105: 102821, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38218172

RESUMEN

Dyslipidemia is frequently observed in polycystic ovarian syndrome (PCOS). Changes in plasma lipid levels potentially alter erythrocyte membrane lipid composition due to lack of inbuilt lipid synthesis machinery. Therefore, development of morphologically altered erythrocytes in PCOS patients with dyslipidemia is expected. However, this has not been established so far. So, we took this opportunity to explore the morphological alterations among dyslipidemic PCO women. We recruited thirty-five dyslipidemic PCOS women (satisfying Rotterdam criteria, without medication) and twenty-five age-matched healthy controls. Scanning electron microscopy revealed a significant increase in the number of stomatocytes, acanthocytes, and echinocytes in the PCO group. PCO group showed a considerable decrease in plasma antioxidant levels. Elevated lipid peroxidation, protein carbonylation, and decreased free thiol group in erythrocyte membrane in PCOS suggest oxidative degradation of the erythrocyte membrane. Elevated intracellular ROS levels, increased methemoglobin formation, and a decrease in NADPH methemoglobin reductase in PCOS also indicate altered physicochemical property of hemoglobin due to oxidative overload. Additionally, these patients exhibit a rise in erythrocyte membrane cholesterol and triglyceride, which promotes the membrane to become less fluidic and less fragile. Thus, these results corroborate a potential role in altering erythrocyte morphology among dyslipidemic PCO women.


Asunto(s)
Dislipidemias , Síndrome del Ovario Poliquístico , Humanos , Femenino , Triglicéridos , Antioxidantes , Eritrocitos , Dislipidemias/complicaciones
15.
Mol Hum Reprod ; 30(3)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38291901

RESUMEN

PCOS is a complex and heterogeneous metabolic disorder that affects 6-20% of women of reproductive age. However, research on phosphorylation modification proteomics in PCOS remains lacking. PCOS can be divided into two groups based on the presence or absence of insulin resistance: PCOS with insulin resistance (PCOS-IR) and PCOS non-insulin resistant (PCOS-NIR). This study focused on the group without insulin resistance. Twenty-one PCOS-NIR and 39 control-NIR (Ctrl-NIR) patients were included in this study. All participants underwent ICSI or IVF-embryo transfer (IVF-ET) treatment in a reproductive center from July 2020 to November 2020. During oocyte retrieval, fresh follicular fluid was aspirated, collected, and sent to the laboratory for analysis of the granulosa cells. A 4D-label-free proteome quantification method was performed in this study; this was used to analyze protein enzymatic peptide fragments by liquid chromatography-mass spectrometry (LC-MS). Bioinformatic analysis was performed on differentially expressed proteins (DEPs) and differentially phosphorylated proteins (DPPs). A total of 713 DEPs were identified between the two groups, including 293 upregulated and 420 downregulated DEPs in the PCOS-NIR group. There were 522 and 159 proteins with increased and decreased phosphorylation, respectively, in the PCOS-NIR group. After analyzing the different phosphorylation modification sites, 933 sites with upregulated and 211 sites with downregulated phosphorylation were found in the PCOS-NIR group. In this study, we describe the quantitative protein expression profiles and phosphorylation-modified protein expression profiles of ovarian granulosa cells from patients with PCOS-NIR, providing a new research perspective for these patients. Further studies are required to elucidate the role of protein phosphorylation in PCOS.


Asunto(s)
Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Humanos , Femenino , Proteómica , Biología Computacional , Células de la Granulosa
16.
Mol Hum Reprod ; 30(5)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38718206

RESUMEN

Paxillin is a ubiquitously expressed adaptor protein integral to focal adhesions, cell motility, and apoptosis. Paxillin has also recently been implicated as a mediator of nongenomic androgen receptor (AR) signaling in prostate cancer and other cells. We sought to investigate the relationship between paxillin and AR in granulosa cells (GCs), where androgen actions, apoptosis, and focal adhesions are of known importance, but where the role of paxillin is understudied. We recently showed that paxillin knockout in mouse GCs increases fertility in older mice. Here, we demonstrate that paxillin knockdown in human granulosa-derived KGN cells, as well as knockout in mouse primary GCs, results in reduced AR protein but not reduced mRNA expression. Further, we find that both AR protein and mRNA half-lives are reduced by approximately one-third in the absence of paxillin, but that cells adapt to chronic loss of paxillin by upregulating AR gene expression. Using co-immunofluorescence and proximity ligation assays, we show that paxillin and AR co-localize at the plasma membrane in GCs in a focal adhesion kinase-dependent way, and that disruption of focal adhesions leads to reduced AR protein level. Our findings suggest that paxillin recruits AR to the GC membrane, where it may be sequestered from proteasomal degradation and poised for nongenomic signaling, as reported in other tissues. To investigate the physiological significance of this in disorders of androgen excess, we tested the effect of GC-specific paxillin knockout in a mouse model of polycystic ovary syndrome (PCOS) induced by chronic postnatal dihydrotestosterone (DHT) exposure. While none of the control mice had estrous cycles, 33% of paxillin knockout mice were cycling, indicating that paxillin deletion may offer partial protection from the negative effects of androgen excess by reducing AR expression. Paxillin-knockout GCs from mice with DHT-induced PCOS also produced more estradiol than GCs from littermate controls. Thus, paxillin may be a novel target in the management of androgen-related disorders in women, such as PCOS.


Asunto(s)
Adhesiones Focales , Células de la Granulosa , Ratones Noqueados , Paxillin , Receptores Androgénicos , Animales , Femenino , Humanos , Ratones , Adhesiones Focales/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Células de la Granulosa/efectos de los fármacos , Paxillin/metabolismo , Paxillin/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Transducción de Señal
17.
Clin Endocrinol (Oxf) ; 100(3): 278-283, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38127445

RESUMEN

OBJECTIVE: Spironolactone (SPL) has been used to manage hyperandrogenic manifestations in women with polycystic ovary syndrome (PCOS), but data on the risk of hyperkalemia in this population are scarce. The aim of this study was to evaluate the incidence of hyperkalemia in women with PCOS using SPL in the long term. DESIGN: Single-centre retrospective study. PATIENTS: Inclusion and analysis of 98 treatment periods in 78 women with PCOS (20 of whom were duplicates, returning after treatment interruption for a mean of 38 months) who received SPL for a minimum of 12 months and had at least three measurements of potassium levels over time. MEASUREMENTS: Clinical and hormonal profiles before and during SPL treatment. RESULTS: Mean age was 29.1 (SD: 9.6) years, and body mass index was 32.2 (SD: 8.1) kg/m². Nine patients had diabetes, and 22 had prediabetes. SPL was used in combination with combined oral contraceptive pills in 55 participants and progestin-only pills/long-acting reversible contraception in 28; metformin was added in 35, and angiotensin-converting enzyme inhibitors/angiotensin receptor blockers in 15. Median SPL dose was 100 (range: 50-150) mg. A total of 327 serum potassium measurements were obtained (84 pre-exposure and 243 postexposure). Four potassium measurements were above the reference range before exposure and 19 during exposure. All potassium measurements above the reference range during follow-up were classified as mild hyperkalemia (5.1-5.5 mEq/L). CONCLUSIONS: The present findings suggest that women with PCOS, without kidney or heart disease, using SPL combined with hormonal contraception for managing clinical hyperandrogenism have a low incidence of hyperkalemia and well-tolerated minor adverse effects.


Asunto(s)
Hiperpotasemia , Síndrome del Ovario Poliquístico , Potasio , Espironolactona , Adulto , Femenino , Humanos , Hirsutismo , Hiperpotasemia/inducido químicamente , Hiperpotasemia/complicaciones , Hiperpotasemia/tratamiento farmacológico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Potasio/sangre , Estudios Retrospectivos , Espironolactona/efectos adversos
18.
Hum Reprod ; 39(8): 1794-1803, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38867472

RESUMEN

STUDY QUESTION: Is resting energy expenditure (REE) altered in women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER: Women with PCOS have a reduction in REE, when corrected for fat-free mass, independent of PCOS clinical phenotypes and BMI categories. WHAT IS KNOWN ALREADY: Obesity is an important issue in women with PCOS, in terms of frequency and pathophysiological implications. It has been hypothesized that obesity may be favoured by alterations in REE, but the studies have been limited and conflicting. STUDY DESIGN, SIZE, DURATION: This case-control study was a comparison of 266 women with PCOS and 51 healthy controls, recruited in the Verona 3P study from 2010 to 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS: Women with PCOS diagnosed by the Rotterdam criteria, with normal thyroid function and no interfering medications, were referred to the outpatient clinic of a tertiary care centre of endocrinology and metabolism for a measurement of REE. Healthy controls were recruited in the same period and submitted to the same procedure. In all subjects, REE was measured by indirect calorimetry and serum androgens were measured by LC-MS/MS. In women with PCOS, insulin sensitivity was assessed using the hyperinsulinemic-euglycemic clamp. MAIN RESULTS AND THE ROLE OF CHANCE: REE was similar in women with PCOS and controls. However, REE corrected for fat-free mass (REE/FFM) was significantly lower in women with PCOS than in controls (31.8 ± 4.0 vs 35.4 ± 3.9 kcal/kgFFM·day, P < 0.001). REE/FFM did not differ between normal-weight, overweight, or obese women with PCOS, and each of these subgroups showed lower REE/FFM values than controls. Reduced REE/FFM values were found in each phenotype of the syndrome. In multiple regression analysis, REE/FFM was independently associated with age and PCOS status, but not with fat mass. In PCOS women, REE/FFM was independently and directly associated with ovarian follicle number. LIMITATIONS, REASONS FOR CAUTION: Limitations of the study are the cross-sectional design, which limits the causal inference of the results, and the unavailability of precise information about lifestyle factors, which may be potential confounders. Further prospective studies are needed to establish the importance of this phenomenon in contributing to the weight excess of PCOS. WIDER IMPLICATIONS OF THE FINDINGS: A reduction of REE could potentially favour weight gain in women with PCOS and possibly contribute to the altered metabolic profile typical of this condition, even counteracting the therapeutic strategies aimed to reduce excess body fat in these women. Nevertheless, the presence of this abnormality in both obese/overweight and normal-weight patients suggests that other factors must play a role in this phenomenon. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by academic grants to PM from the University of Verona (FUR 2010-2022). All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Metabolismo Energético , Obesidad , Síndrome del Ovario Poliquístico , Humanos , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/fisiopatología , Síndrome del Ovario Poliquístico/complicaciones , Femenino , Adulto , Estudios de Casos y Controles , Obesidad/metabolismo , Obesidad/complicaciones , Obesidad/fisiopatología , Adulto Joven , Resistencia a la Insulina , Índice de Masa Corporal , Metabolismo Basal , Calorimetría Indirecta
19.
Hum Reprod ; 39(1): 108-118, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38011904

RESUMEN

STUDY QUESTION: What is the current burden of infertility attributable to PCOS at global, regional, and national levels by age and socio-demographic index (SDI) across 21 regions and 204 countries and territories? SUMMARY ANSWER: The burden of infertility attributable to PCOS increased from 6.00 million prevalent cases in 1990 to 12.13 million in 2019 globally and increased sharply in most regions and nations. WHAT IS KNOWN ALREADY: PCOS is the most common cause of anovulatory infertility, affecting up to 80% of women with anovulation. No comprehensive and detailed epidemiological estimates of infertility attributable to PCOS in reproductive women aged 15-49 years by age and SDI, at the global, regional, and national level, have been reported. STUDY DESIGN, SIZE, DURATION: An age- and SDI-stratified systematic analysis of the prevalence and years lived with disability (YLD) of infertility attributable to PCOS across 21 regions and 204 countries and territories from 1990 to 2019 has been performed. PARTICIPANTS/MATERIALS, SETTING, METHODS: The prevalence and YLD of female infertility attributable to PCOS in reproductive women aged 15-49 years from 1990 to 2019 were retrieved directly from the Global Burden of Diseases 2019. The number, rates per 100 000 persons, and average annual percentage changes (AAPCs) of prevalence and YLD were estimated at the global, regional, and national levels. MAIN RESULTS AND THE ROLE OF CHANCE: Globally, the prevalent cases of infertility attributable to PCOS among women of reproductive age (15-49 years) doubled from 1990 to 2019, with 6.00 million prevalent cases in 1900 and 12.13 million in 2019. The global age-standardized prevalence rates (ASPRs) of infertility attributable to PCOS were 223.50/100 000 persons in 1990 and 308.25/100 000 persons in 2019. At global level, the YLDs of infertility attributable to PCOS increased by 98.0% from 35.20 thousand in 1990 to 69.70 thousand in 2019. The burden of infertility attributable to PCOS in the high SDI region was significantly higher than that in the other four SDI regions. The greatest annual increases in rates of ASPR and age-standardized YLD rate were observed in the middle SDI region (AAPC 1.96 [95% CI 1.87-2.06], 1.94 [1.87-2.00], respectively) and the low-middle SDI region (AAPC 1.96 [1.90-2.03], 1.90 [1.85-1.94], respectively). The regional highest ASPR and the age-standardized YLD rate of infertility were observed in High-income Asia Pacific. The national highest ASPR and the age-standardized YLD rate of infertility were observed in Italy. Positive associations were observed between these burden estimates and the SDI level (all P < 0.001). LIMITATIONS, REASONS FOR CAUTION: Although the Global Burden of Diseases 2019 has tried its best to collect all available data, some countries have limited data, which may result in an underestimation of the burden of infertility attributable to PCOS. The diagnostic criteria of PCOS are constantly changing, which may induce bias in infertility attributable to PCOS. No information on the PCOS phenotype is provided in the Global Burden of Diseases 2019, so we cannot estimate the infertility attributable to a specific PCOS phenotype. Detection bias would lead to a higher prevalence of PCOS and infertility attributable to PCOS in developed countries with well-established medical systems and greater willingness of the populace to seek medical attention. Thus, health resource allocation for infertility attributable to PCOS in low-prevalence areas should not be ignored. WIDER IMPLICATIONS OF THE FINDINGS: The global burden of infertility attributable to PCOS increased sharply from 1990 to 2019. Effective health interventions and efficient preventative and managerial strategies should be established to reduce the burden of infertility attributable to PCOS. Weight control is suggested to reduce the burden of infertility attributable to PCOS, especially in the high SDI region. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the National Key Research and Development Program of China (grant number, 2022YFC2704100) and the National Natural Science Foundation of China (Nos 82001498 and 82371648). The authors declare no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Infertilidad , Síndrome del Ovario Poliquístico , Humanos , Femenino , Años de Vida Ajustados por Calidad de Vida , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/epidemiología , Carga Global de Enfermedades , Prevalencia , Salud Global
20.
Hum Reprod ; 39(3): 586-594, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38177084

RESUMEN

STUDY QUESTION: Do ongoing pregnancy rates (OPRs) differ in predicted hyperresponders undergoing ART after IVM of oocytes compared with conventional ovarian stimulation (OS) for IVF/ICSI? SUMMARY ANSWER: One cycle of IVM is non-inferior to one cycle of OS in women with serum anti-Müllerian hormone (AMH) levels ≥10 ng/ml. WHAT IS KNOWN ALREADY: Women with high antral follicle count and elevated serum AMH levels, indicating an increased functional ovarian reserve, are prone to hyperresponse during ART treatment. To avoid iatrogenic complications of OS, IVM has been proposed as a mild-approach alternative treatment in predicted hyperresponders, including women with polycystic ovary syndrome (PCOS) who are eligible for ART. To date, inferior pregnancy rates from IVM compared to OS have hampered the uptake of IVM by ART clinics. However, it is unclear whether the efficiency gap between IVM and OS may differ depending on the extent of AMH elevation. STUDY DESIGN, SIZE, DURATION: This study is a retrospective cohort analysis of clinical and laboratory data from the first completed highly purified hMG (HP-hMG) primed, non-hCG-triggered IVM or OS (FSH or HP-hMG stimulation in a GnRH antagonist protocol) cycle with ICSI in predicted hyperresponders ≤36 years of age at a tertiary referral university hospital. A total of 1707 cycles were included between January 2016 and June 2022. PARTICIPANTS/MATERIALS, SETTING, METHODS: Predicted hyperresponse was defined as a serum AMH level ≥3.25 ng/ml (Elecsys® AMH, Roche Diagnostics). The primary outcome was cumulative ongoing pregnancy rate assessed 10-11 weeks after embryo transfer (ET). The predefined non-inferiority limit was -10.0%. The analysis was adjusted for AMH strata. Time-to-pregnancy, defined as the number of ET cycles until ongoing pregnancy was achieved, was a secondary outcome. Statistical analysis was performed using a multivariable regression model controlling for potential confounders. MAIN RESULTS AND THE ROLE OF CHANCE: Data from 463 IVM cycles were compared with those from 1244 OS cycles. Women in the IVM group more often had a diagnosis of Rotterdam PCOS (434/463, 93.7%) compared to those undergoing OS (522/1193, 43.8%), were significantly younger (29.5 years versus 30.5 years, P ≤ 0.001), had a higher BMI (25.7 kg/m2 versus 25.1 kg/m2, P ≤ 0.01) and higher AMH (11.6 ng/ml versus 5.3 ng/ml, P ≤ 0.001). Although IVM cycles yielded more cumulus-oocyte complexes (COCs) (24.5 versus 15.0 COC, P ≤ 0.001), both groups had similar numbers of mature oocytes (metaphase II (MII)) (11.9 MII versus 10.6 MII, P = 0.9). In the entire cohort, non-adjusted cumulative OPR from IVM was significantly lower (198/463, 42.8%) compared to OS (794/1244, 63.8%), P ≤ 0.001. When analysing OPR across different serum AMH strata, cumulative OPR in both groups converged with increasing serum AMH, and OPR from IVM was non-inferior compared to OS from serum AMH levels >10 ng/ml onwards (113/221, 51.1% (IVM); 29/48, 60.4% (OS)). The number of ETs needed to reach an ongoing pregnancy was comparable in both the IVM and the OS group (1.6 versus 1.5 ET's, P = 0.44). Multivariable regression analysis adjusting for ART type, age, BMI, oocyte number, and PCOS phenotype showed that the number of COCs was the only parameter associated with OPR in predicted hyperresponders with a serum AMH >10 ng/ml. LIMITATIONS, REASONS FOR CAUTION: These data should be interpreted with caution as the retrospective nature of the study holds the possibility of unmeasured confounding factors. WIDER IMPLICATIONS OF THE FINDINGS: Among subfertile women who are eligible for ART, IVM, and OS resulted in comparable reproductive outcomes in a subset of women with a serum AMH ≥10 ng/ml. These findings should be corroborated by a randomised controlled trial (RCT) comparing both treatments in selected patients with elevated AMH. STUDY FUNDING/COMPETING INTEREST(S): There was no external funding for this study. P.D. has been consultant to Merck Healthcare KGaA (Darmstadt, Germany) from April 2021 till June 2023 and is a Merck employee (Medical Director, Global Medical Affairs Fertility) with Merck Healthcare KGAaA (Darmstadt, Germany) since July 2023. He declares honoraria for lecturing from Merck KGaA, MSD, Organon, and Ferring. The remaining authors declared no conflict of interest pertaining to this study. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Técnicas de Maduración In Vitro de los Oocitos , Síndrome del Ovario Poliquístico , Femenino , Humanos , Embarazo , Hormona Antimülleriana , Oocitos , Síndrome del Ovario Poliquístico/terapia , Inyecciones de Esperma Intracitoplasmáticas , Estudios Retrospectivos , Adulto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA