Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 705
Filtrar
1.
Obesity (Silver Spring) ; 32(4): 678-690, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38439205

RESUMO

OBJECTIVE: Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, insulin resistance, and hepatic steatosis (HS). Because dietary essential amino acid (EAA) supplementation has been shown to decrease HS in various populations, this study's objective was to determine whether supplementation would decrease HS in PCOS. METHODS: A randomized, double-blind, crossover, placebo-controlled trial was conducted in 21 adolescents with PCOS (BMI 37.3 ± 6.5 kg/m2, age 15.6 ± 1.3 years). Liver fat, very low-density lipoprotein (VLDL) lipogenesis, and triacylglycerol (TG) metabolism were measured following each 28-day phase of placebo or EAA. RESULTS: Compared to placebo, EAA was associated with no difference in body weight (p = 0.673). Two markers of liver health improved: HS was lower (-0.8% absolute, -7.5% relative reduction, p = 0.013), as was plasma aspartate aminotransferase (AST) (-8%, p = 0.004). Plasma TG (-9%, p = 0.015) and VLDL-TG (-21%, p = 0.031) were reduced as well. VLDL-TG palmitate derived from lipogenesis was not different between the phases, nor was insulin sensitivity (p > 0.400 for both). Surprisingly, during the EAA phase, participants reported consuming fewer carbohydrates (p = 0.038) and total sugars (p = 0.046). CONCLUSIONS: Similar to studies in older adults, short-term EAA supplementation in adolescents resulted in significantly lower liver fat, AST, and plasma lipids and thus may prove to be an effective treatment in this population. Additional research is needed to elucidate the mechanisms for these effects.


Assuntos
Fígado Gorduroso , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Adolescente , Feminino , Humanos , Hiperandrogenismo/complicações , Insulina , Lipoproteínas VLDL , Obesidade/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/complicações
2.
Medicina (Kaunas) ; 60(2)2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38399531

RESUMO

Polycystic ovary syndrome (PCOS) manifests a multifactorial pathology characterized by polycystic ovaries, menstrual cycle disorders, varying degrees of hyperandrogenism, and an ad-verse metabolic risk profile. The position of hyperandrogenism in this syndrome has been extensively studied. A multitude of mechanisms place it in the position of cause but also of consequence; therefore, ongoing research efforts are focused on identifying medications that can effectively reduce levels of androgens in women with PCOS. Moreover, lipid abnormalities are common in this population, with up to 70% of patients having dyslipidemia. Statins may have potential therapeutic benefits for women with PCOS, as they have been shown to improve insulin resistance and reduce the risk of cardiovascular disease. In addition, their role in accelerated steroidogenesis by limiting one source of cholesterol, influencing enzymatic activity, and providing several other beneficial mechanisms is widely investigated. This review aimed to provide a comprehensive overview of the pathogenesis of androgen excess and dyslipidemia in PCOS, as well as the therapeutic potential of statins.


Assuntos
Dislipidemias , Inibidores de Hidroximetilglutaril-CoA Redutases , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Humanos , Feminino , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Hiperandrogenismo/complicações , Hiperandrogenismo/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Dislipidemias/complicações , Dislipidemias/tratamento farmacológico
3.
Expert Rev Endocrinol Metab ; 19(2): 111-128, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38205927

RESUMO

INTRODUCTION: Hyperandrogenism is a clinical state consequent to excess androgen production by the ovary, adrenals, or increased peripheral conversion of androgens. The varied manifestations of hyperandrogenism include seborrhea, acne, infertility, hirsutism, or overt virilization of which adult female acne, hirsutism, and female pattern hair loss are of clinical relevance to dermatologists. AREAS COVERED: We limited our narrative review to literature published during period from 1 January 1985 to Dec 2022 and searched PubMed/MEDLINE, Web of Science (WOS), Scopus, and Embase databases with main search keywords were 'Hyperandrogenism,' 'Female,' 'Biochemical,' 'Dermatological', and 'Dermatology.' We detail the common etiological causes, nuances in interpretation of biochemical tests and imaging tools, followed by an algorithmic approach which can help avoid extensive tests and diagnose the common causes of hyperandrogenism. EXPERT OPINION: Based on current data, total testosterone, sex hormone binding globulin, DHEAS, prolactin, free androgen index, and peripheral androgenic metabolites like 3-alpha diol and androsterone glucuronide are ideal tests though not all are required in all patients. Abnormalities in these biochemical investigations may require radiological examination for further clarification. Total testosterone levels can help delineate broadly the varied causes of hyperandrogenism. Serum AMH could be used for defining PCOM in adults.


Assuntos
Acne Vulgar , Hiperandrogenismo , Adulto , Humanos , Feminino , Hirsutismo/diagnóstico , Hirsutismo/etiologia , Hiperandrogenismo/complicações , Hiperandrogenismo/diagnóstico , Androgênios , Dermatologistas , Testosterona/metabolismo , Alopecia/diagnóstico , Alopecia/etiologia , Acne Vulgar/diagnóstico , Acne Vulgar/etiologia
4.
Gynecol Obstet Invest ; 89(1): 22-30, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38194939

RESUMO

INTRODUCTION: Polycystic ovary syndrome (PCOS) is a common hormonal disorder among young women, correlated with hyperandrogenism. Among the symptoms of PCOS, vocal alterations are quite unknown. Dysphonia may be related to hyperandrogenism, and there is no consensus about its prevalence and the severity of vocal disorders, which can cause noticeable discomfort. METHODS: A systematic review of the literature was conducted. Four studies on PCOS that evaluated the phonatory system were included for a total of 174 patients (96 PCOS, 78 controls), and a meta-analysis on comparable data was performed. RESULTS: Four studies evaluated parameters related to vocal symptomatology, altered audiometric examination, and findings at the laryngoscopy in patients affected by PCOS versus controls. Although the individual studies showed increased incidence of alterations and a tendency to develop speech fatigue in women with PCOS, when the results of studies were pulled in meta-analysis, the overall difference was not statistically significant. The studies themselves were very different from each other; therefore, it is hard to draw any firm conclusions. DISCUSSION: The aim of this study was to assess the prevalence of vocal alterations, the correlation with hyperandrogenism, the quality of life, and the voice changes after starting a therapy for PCOS. The present meta-analysis failed to find any difference in terms of PCOS and control cohort. However, the lack of high-quality studies makes it difficult to draw firm conclusions. New and larger studies or big population program data are therefore warranted.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/epidemiologia , Hiperandrogenismo/complicações , Hiperandrogenismo/epidemiologia , Qualidade de Vida
5.
Biomol Concepts ; 15(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38242137

RESUMO

In the past two decades, oxidative stress (OS) has drawn a lot of interest due to the revelation that individuals with many persistent disorders including diabetes, polycystic ovarian syndrome (PCOS), cardiovascular, and other disorders often have aberrant oxidation statuses. OS has a close interplay with PCOS features such as insulin resistance, hyperandrogenism, and chronic inflammation; there is a belief that OS might contribute to the development of PCOS. PCOS is currently recognized as not only one of the most prevalent endocrine disorders but also a significant contributor to female infertility, affecting a considerable proportion of women globally. Therefore, the understanding of the relationship between OS and PCOS is crucial to the development of therapeutic and preventive strategies for PCOS. Moreover, the mechanistic study of intracellular reactive oxygen species/ reactive nitrogen species formation and its possible interaction with women's reproductive health is required, which includes complex enzymatic and non-enzymatic antioxidant systems. Apart from that, our current review includes possible regulation of the pathogenesis of OS. A change in lifestyle, including physical activity, various supplements that boost antioxidant levels, particularly vitamins, and the usage of medicinal herbs, is thought to be the best way to combat this occurrence of OS and improve the pathophysiologic conditions associated with PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/terapia , Síndrome do Ovário Policístico/complicações , Antioxidantes/uso terapêutico , Hiperandrogenismo/complicações , Estresse Oxidativo
6.
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
7.
Artigo em Inglês | MEDLINE | ID: mdl-37664890

RESUMO

This study discusses the relationship between Polycystic Ovary Syndrome (PCOS) and diabetes in women, which has become increasingly prevalent due to changing lifestyles and environmental factors. The characteristic that distinguishes women with PCOS is hyperandrogenism which results from abnormal ovarian or adrenal function, which leads to the overproduction of androgens. Excessive androgens in women increase the risk of Type 2 diabetes (T2D) and insulin resistance (IR). Nowadays, diabetes affects people of all ages and is linked to factors such as lifestyle, genetics, stress, and aging. Diabetes, the uncontrolled high blood sugar level can potentially harm kidneys, nerves, eyes, and other organs and there is no cure, making it a concerning disease in developing nations. This research tried to submit the evidence through feature-wise correlation analyses between PCOS and diabetes. Hence, this model utilized the Exploratory Data Analysis (EDA) and the Elbow clustering algorithms for the experimental purpose in which the EDA deeply analyzed the features of PCOS and diabetes and recorded a positive correlation of 95%. The Elbow clustering technique is employed for verifying the correlations identified through EDA. Although limited research exists on this specific disease, this work provides potential evidence for the research community by evaluating the clustering results using Silhouette Score, Calinski-Harabasz Index, and Davies-Bouldin Index.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Diabetes Mellitus Tipo 2/etiologia , Fatores de Risco , Hiperandrogenismo/complicações , Resistência à Insulina/fisiologia
8.
J Pediatr Adolesc Gynecol ; 37(1): 51-55, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37696388

RESUMO

STUDY OBJECTIVE: To determine the incidence of polycystic ovarian syndrome (PCOS) and hyperandrogenism among adolescent transmasculine patients presenting to a tertiary care referral center for gender-affirming care METHODS: This was a retrospective study of adolescent transmasculine patients presenting to Cleveland Clinic for gender-affirming hormone therapy. The diagnostic criteria were adolescent-specific as defined by the international evidence-based guideline for PCOS management and included oligomenorrhea and/or anovulation with clinical and/or biochemical hyperandrogenism after exclusion of other androgen excess disorders. RESULTS: The described transgender population had a prevalence of PCOS of 23.8%. The transmasculine patients who met the criteria for PCOS had both higher levels of androgens and higher body mass indexes when compared with the patients without PCOS. Additionally, the patients with PCOS had higher rates of dyslipidemia. CONCLUSION: The prevalence of PCOS among transmasculine patients may be higher compared with the general population. Transmasculine patients with PCOS should be counseled regarding the long-term health implications associated with PCOS and screened appropriately to minimize risks.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Adolescente , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/epidemiologia , Síndrome do Ovário Policístico/diagnóstico , Hiperandrogenismo/complicações , Estudos Retrospectivos , Prevalência
9.
J Pediatr Adolesc Gynecol ; 37(2): 217-219, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38110028

RESUMO

Ovarian tumors are rare in children; however, their incidence increases with age. Of these ovarian tumors, Leydig cell tumors are some of the rarest, accounting for less than 0.1% of all ovarian tumors across all ages. Leydig cell tumors predominantly occur in postmenopausal women and are characterized by nodular proliferation of Leydig cells in the ovarian hilum with intracytoplasmic Reinke crystals. These tumors secrete androgens, which can disrupt ovarian function, clinically presenting with abnormal uterine bleeding and virilization. Although they are generally benign, current recommendations are for treatment with a unilateral salpingo-oophorectomy. In adolescents, hyperandrogenism is most commonly caused by polycystic ovarian syndrome (PCOS); however, the differential for hyperandrogenism is broad. We present a case of a 15-year-old girl with a history of primary amenorrhea who presented with a Leydig cell tumor associated with recurrent ovarian torsion and virilization. This case reviews the challenges with diagnosis, management, and future implications of a rare androgen-secreting tumor in young patients.


Assuntos
Hiperandrogenismo , Tumor de Células de Leydig , Neoplasias Ovarianas , Masculino , Criança , Humanos , Feminino , Adolescente , Tumor de Células de Leydig/complicações , Tumor de Células de Leydig/cirurgia , Tumor de Células de Leydig/diagnóstico , Hiperandrogenismo/complicações , Virilismo/etiologia , Neoplasias Ovarianas/complicações , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/cirurgia , Androgênios
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(9): 1425-1431, 2023.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38044655

RESUMO

Hyperandrogenism-insulin resistance-acanthosis nigricans (HAIR-AN) syndrome is a special and rare subtype of polycystic ovarian syndrome. It can lead to hyperandrogenism (HA), insulin resistance (IR), and acanthosis nigricans (AN) accompanied by acne, hirutism, irregular menstruation, and other androgen excess symptoms. A case of pediatric HAIR-AN syndrome with severe AN was admitted to the Department of Endocrinology, China-Japan Friendship Hospital. The patient's clinical manifestations, laboratory data, imaging features, and gene sequencing were analyzed, and the patient was diagnosed with pediatric HAIR-AN syndrome. Obesity, IR, hyperglycemia, menstrual disorder, and AN were significantly improved after treating with metformin and liraglutide. HAIR-AN syndrome occurs in various forms. When the patient appears unexplained acanthosis nigricans and menstrual disorders, the disease should be considered possible. Early diagnosis and symptomatic supportive treatment can improve the quality of life.


Assuntos
Acantose Nigricans , Hiperandrogenismo , Resistência à Insulina , Síndrome Metabólica , Metformina , Síndrome do Ovário Policístico , Feminino , Criança , Humanos , Hiperandrogenismo/complicações , Hiperandrogenismo/diagnóstico , Hiperandrogenismo/genética , Acantose Nigricans/complicações , Acantose Nigricans/diagnóstico , Acantose Nigricans/genética , Liraglutida/uso terapêutico , Metformina/uso terapêutico , Qualidade de Vida , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/complicações , Síndrome Metabólica/complicações , Síndrome Metabólica/tratamento farmacológico
11.
Curr Atheroscler Rep ; 25(12): 1003-1011, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38048007

RESUMO

PURPOSE OF REVIEW: Characterize the risk of cardiovascular disease (CVD) in individuals with polycystic ovarian syndrome (PCOS). Review the pathophysiological pathways that confers CVD risk in individuals with PCOS and interventions to reduce CVD risk. RECENT FINDINGS: PCOS is a complex syndrome characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovaries that has metabolic and cardiovascular implications. Intrinsic hormonal dysregulation and chronic low-grade inflammation play an important role in the progression of atherosclerosis in young premenopausal individuals and development of CVD independently of associated traditional risk factors. Management with metformin reduces CVD risk by reducing atherosclerosis progression. PCOS is an important CVD risk factor among individuals of reproductive age. Early detection and interventions are needed to mitigate development of CVD.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/metabolismo , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Fatores de Risco , Hiperandrogenismo/complicações , Hiperandrogenismo/diagnóstico , Inflamação/complicações , Aterosclerose/complicações
12.
Medicine (Baltimore) ; 102(50): e36075, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38115365

RESUMO

Polycystic ovary syndrome (PCOS) is a complex disorde7r influenced by genetic, neuroendocrine, metabolic, environmental, and lifestyle factors. This paper delves into the increasingly recognized role of gut microbiota dysbiosis in the onset and progression of PCOS. Utilizing advances in next-generation sequencing and metabolomics, the research examines the intricate interaction between the gut microbiota and the central nervous system via the gut-brain axis. The paper highlights how disruptions in gut microbiota contribute significantly to PCOS by modulating the release of gut-brain peptides and activating inflammatory pathways. Through such mechanisms, gut microbiota dysbiosis is implicated in hyperandrogenism, insulin resistance, chronic inflammation, and metabolic disorders associated with PCOS. While the relationship between gut microbiota and PCOS has begun to be elucidated, this paper underscores the need for further research to identify specific bacterial strains and their metabolic byproducts as potential therapeutic targets. Therefore, comprehensive studies are urgently needed to understand and fundamentally treat the pathophysiological processes of PCOS, offering valuable insights for future treatment and prevention strategies.


Assuntos
Microbioma Gastrointestinal , Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/tratamento farmacológico , Microbioma Gastrointestinal/fisiologia , Disbiose , Hiperandrogenismo/complicações , Resistência à Insulina/genética
13.
Obstet Gynecol Clin North Am ; 50(4): 695-705, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37914488

RESUMO

Polycystic ovary syndrome (PCOS) is a complex syndrome that affects menstrual regularity, causes hyperandrogenism, increases the risk of metabolic dysfunction and infertility, and is associated with higher rates of mental health disorders. The symptoms of PCOS are unique to each individual and will evolve throughout their reproductive lifespan and beyond. Thus, care should be personalized and provided by an appropriate team of multidisciplinary physicians and clinicians, such as dieticians and psychologists.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/terapia , Hiperandrogenismo/terapia , Hiperandrogenismo/complicações
14.
Probl Endokrinol (Mosk) ; 69(5): 107-114, 2023 Nov 12.
Artigo em Russo | MEDLINE | ID: mdl-37968958

RESUMO

Polycystic ovary syndrome (PCOS) is one of the most pressing problems in endocrine gynecology. The main signs of the disease are hyperandrogenism, menstrual and/or ovulatory dysfunction, and polycystic ovarian structure according to ultrasound. Women with PCOS are at risk for developing metabolic syndrome, type 2 diabetes, cardiovascular disease, and endometrial cancer. In this connection, the pathogenetic mechanisms of the occurrence of this syndrome are continuously studied and new methods of treatment are being sought. PCOS is characterized by a wide range of various disorders of the neuroendocrine regulation of the reproductive system. The main focus of the review is aimed at summarizing information about the etiological role of neuropeptides and neurotransmitters, such as phoenixin, galanins, orexins, GABA, in the pathophysiology of PCOS and about the possibility of their use for diagnostic and therapeutic purposes. In recent decades, the interest of scientists has been focused on the study of KNDy neurons, because it is the kisspeptin synthesized by them that is one of the main regulators of the hypothalamic-pituitary-ovarian axis. This article discusses data on the significance of KNDy neurons in the pathogenesis of the syndrome. Information is provided on the effect of elevated levels of androgens and anti-Müllerian hormone on GnRH neurons. Also analyzed are studies on functional and structural disorders in the hypothalamus in PCOS. Literature search was carried out in national (eLibrary, CyberLeninka.ru) and international (PubMed, Cochrane Library) databases in Russian and English. The priority was free access to the full text of articles. The choice of sources was prioritized for the period from 2018 to 2023.However, taking into account the insufficient knowledge of the chosen topic, the choice of sources dates back to 1998.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Diabetes Mellitus Tipo 2/complicações , Hiperandrogenismo/complicações , Androgênios
15.
Life Sci ; 335: 122280, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37981226

RESUMO

Polycystic Ovary Syndrome (PCOS) and osteoporosis, though seemingly unrelated, exhibit intricate connections influenced by genetic and epigenetic factors. PCOS, characterized by elevated androgen levels, insulin resistance, and increased body weight, has historically been considered protective against bone fragility disorders. However, emerging research suggests that chronic inflammation, prevalent in PCOS, can adversely affect bone health. Studies have demonstrated variable bone mineral density loss in PCOS, often associated with leptin resistance and hyperinsulinemia. Key genes such as INS, IGF1, CTNNB1, AKT1, and STAT3 play pivotal roles in the complex interplay between PCOS and osteoporosis, influencing insulin signaling, oxidative stress, and inflammatory pathways. Oxidative stress, a prominent element in PCOS, can lead to osteoporosis through hormonal imbalances, chronic inflammation, insulin resistance, and lifestyle factors. The insulin signaling pathway also significantly impacts both conditions by contributing to hormonal imbalances and bone health alterations. This intricate network of genetic and epigenetic factors underscores the need for a deeper understanding of their interrelationships. Thus, this review elucidates the multifaceted genetic, epigenetic, and inflammatory connections between PCOS and osteoporosis, highlighting their implications for bone health management in individuals with PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Osteoporose , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Hiperandrogenismo/complicações , Insulina , Osteoporose/genética , Inflamação/complicações
16.
BMJ Open ; 13(11): e073619, 2023 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-37989369

RESUMO

OBJECTIVE: Serum prokineticin-1 (s-PROK1) in the second and third trimester of pregnancy is positively correlated to preeclampsia, intrauterine growth restriction (IUGR) and preterm delivery. Women with polycystic ovary syndrome (PCOS) are prone to these adverse pregnancy outcomes. However, the contribution of PROK1 to the development of pregnancy complications and the effect of metformin and hyperandrogenism on s-PROK1 in PCOS have not been studied previously. DESIGN: This work is a post hoc analysis of two prospective, randomised, placebo-controlled trials. SETTING: Pregnant women with PCOS were included from 11 study centres in Norway. PARTICIPANTS: From 313 women, 264 participated in the present study after exclusions due to dropouts or insufficient serum samples. INTERVENTION: Women with PCOS were randomly administered with metformin or placebo, from first trimester to delivery. PRIMARY AND SECONDARY OUTCOME MEASURES: s-PROK1 was analysed using ELISA at gestational week 19 and related to pregnancy complications, fasting insulin levels, homoeostatic model assessment for insulin resistance (HOMA-IR), testosterone, or androstenedione levels, metformin use, PCOS phenotype and hyperandrogenism. RESULTS: Maternal s-PROK1 in the second trimester did not predict pregnancy-induced hypertension, pre-eclampsia or late miscarriage/preterm delivery in women with PCOS. However, s-PROK1 was lower in women who used metformin before inclusion, both in those randomised to metformin and to placebo, compared with those who did not. s-PROK1 was also lower in those who used metformin both at conception and during pregnancy compared with those who used metformin from inclusion or did not use metformin at all. s-PROK1 was lower in hyperandrogenic compared with normo-androgenic women with PCOS. CONCLUSIONS: Maternal s-PROK1 in the second trimester did not predict pregnancy complications in PCOS. Those who used metformin at conception and/or during pregnancy had lower s-PROK1. PCOS women with hyperandrogenism exhibited lower s-PROK1 compared with normo-adrogenic phenotypes. TRIAL REGISTRATION NUMBER: NCT03259919 and NCT00159536.


Assuntos
Hormônios Gastrointestinais , Hiperandrogenismo , Metformina , Síndrome do Ovário Policístico , Pré-Eclâmpsia , Complicações na Gravidez , Nascimento Prematuro , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Recém-Nascido , Feminino , Gravidez , Humanos , Metformina/uso terapêutico , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Hiperandrogenismo/induzido quimicamente , Hiperandrogenismo/complicações , Hiperandrogenismo/tratamento farmacológico , Estudos Prospectivos , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/induzido quimicamente , Pré-Eclâmpsia/tratamento farmacológico , Hormônios Gastrointestinais/uso terapêutico
17.
Epigenetics ; 18(1): 2282319, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37992405

RESUMO

Research suggests that polycystic ovary syndrome (PCOS) traits (e.g., hyperandrogenism) may create a suboptimal intrauterine environment and induce epigenetic modifications. Therefore, we assessed the associations of PCOS traits with neonatal DNA methylation (DNAm) using two independent cohorts. DNAm was measured in both cohorts using the Infinium MethylationEPIC array. Multivariable robust linear regression was used to determine associations of maternal PCOS exposure or preconception testosterone with methylation ß-values at each CpG probe and corrected for multiple testing by false-discovery rate (FDR). In the birth cohort, 12% (102/849) had a PCOS diagnosis (8.1% PCOS without hirsutism; 3.9% PCOS with hirsutism). Infants exposed to maternal PCOS with hirsutism compared to no PCOS had differential DNAm at cg02372539 [ß(SE): -0.080 (0.010); FDR p = 0.009], cg08471713 [ß(SE):0.077 (0.014); FDR p = 0.016] and cg17897916 [ß(SE):0.050 (0.009); FDR p = 0.009] with adjustment for maternal characteristics including pre-pregnancy BMI. PCOS with hirsutism was also associated with 8 differentially methylated regions (DMRs). PCOS without hirsutism was not associated with individual CpGs. In an independent preconception cohort, total testosterone concentrations were associated with 3 DMRs but not with individual CpGs, though the top quartile of testosterone compared to the lowest was marginally associated with increased DNAm at cg21472377 near an uncharacterized locus (FDR p = 0.09). Examination of these probes and DMRs indicate they may be under foetal genetic control. Overall, we found several associations among newborns exposed to PCOS, specifically when hirsutism was reported, and among newborns of women with relatively higher testosterone around conception.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Gravidez , Lactente , Humanos , Recém-Nascido , Feminino , Síndrome do Ovário Policístico/genética , Hirsutismo/genética , Hirsutismo/complicações , Hirsutismo/diagnóstico , Metilação de DNA , Hiperandrogenismo/complicações , Hiperandrogenismo/diagnóstico , Testosterona
18.
Clinics (Sao Paulo) ; 78: 100310, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38008036

RESUMO

PCOS is an endocrine disorder characterized by chronic anovulation, hyperandrogenism, and polycystic ovaries. Its etiology is uncertain. It is debated whether BPA would be a component of the environmental factor in the etiology of PCOS. Contamination by BPA can occur from food packaging (exposure during the diet) and through skin absorption and/or inhalation. It can be transferred to the fetus via the placenta or to the infant via breast milk, and it can be found in follicular fluid, fetal serum, and amniotic fluid. The phenolic structure of BPA allows it to interact with Estrogen Receptors (ERs) through genomic signaling, in which BPA binds to nuclear ERα or Erß, or through nongenomic signaling by binding to membrane ERs, prompting a rapid and intense response. With daily and constant exposure, BPA's tendency to bioaccumulate and its ability to activate nongenomic signaling pathways can alter women's metabolic and reproductive function, leading to hyperandrogenism, insulin resistance, obesity, atherogenic dyslipidemia, chronic inflammatory state, and anovulation and favoring PCOS. The harmful changes caused by BPA can be passed on to future generations without the need for additional exposure because of epigenetic modifications. Not only high BPA levels can produce harmful effects, but at low levels, BPA may be harmful when exposure occurs during the most vulnerable periods, such as the fetal and neonatal periods, as well as during the prepubertal age causing an early accumulation of BPA in the body. Learning how BPA participates in the pathogenesis of PCOS poses a challenge and further studies should be conducted.


Assuntos
Anovulação , Hiperandrogenismo , Síndrome do Ovário Policístico , Gravidez , Recém-Nascido , Feminino , Humanos , Síndrome do Ovário Policístico/induzido quimicamente , Hiperandrogenismo/complicações , Anovulação/complicações , Fenóis/toxicidade
19.
Gynecol Endocrinol ; 39(1): 2276167, 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37931646

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) was known as the common endocrine disease in women, featured as hyperandrogenism, ovulation disorders, etc. Fat mass and obesity-associated protein (FTO), a m6A demethylase, is abnormal in the occurrence of ovarian diseases. However, the mechanism of FTO in the pathogenesis of PCOS is still unclear. METHODS: The level of FTO in clinical samples, PCOS rat with hyperandrogenism and granulosa cells (GCs) lines effected by DHT were investigated by ELISA, qRT-PCR, WB, and IHC, while m6A RNA methylation level was studied by m6A Colorimetric and androgen level was tested through ELISA. Changes in steroid hormone synthetase and androgen receptor (AR)/prostate-specific antigen (PSA) levels in vitro were visualized by WB after transient transfection silenced FTO. The effect of DHT combined with FTO inhibitor meclofenamic acid (MA) on FTO, AR/PSA, and AKT phosphorylation were also demonstrated by WB. The co-localization of FTO and AR in KGN cells was analyzed by confocal microscopy, and the physiological interaction between FTO and AR was studied by Co-IP assay. The effect of FTO-specific inhibitor MA, AKT phosphorylation inhibitor LY294002, and the combined them on GCs proliferation and cell cycle were evaluated by drug combination index, EDU assay, and flow cytometry analysis. RESULTS: FTO expression was upregulated in follicular fluid and GCs in PCOS patients clinically. The high FTO expression in patients was negative with the level of m6A, but positive with the level of androgen. The upregulation of FTO was accompanied with a decrease in the level of m6A in PCOS rat with hyperandrogenism. Dihydrotestosterone (DHT) promoted the FTO expression and inhibited m6A content as a dose-dependent way in vitro. In contrast, suppression of FTO with siRNA attenuated the expression of steroid hormone synthetase such as CYP11A1, CYP17A1, HSD11B1, HSD3B2 except CYP19A1 synthetase, ultimately inducing the decrease of androgen level. Suppression of FTO also decreased the biological activity of androgen through downregulation AR/PSA. MA treatment as the specific FTO antagonist decreased cell survival in time- and dose-dependent way in GCs lines. Correspondingly, MA treatment decreased the expression of FTO, AR/PSA expression, and AKT phosphorylation in the presence of DHT stimulation. Additionally, we also speculate there is a potential relation between FTO and AR according to FTO was co-localized and interacted with AR in KGN cells. Compared with AKT phosphorylation inhibitor LY294002 or MA alone, LY294002 combined with MA synergistically inhibited cell survival and increased G2/M phase arrest in GC line. CONCLUSIONS: We first evaluated the correlation of FTO and m6A in PCOS clinically, and further explored the mechanism between FTO and hyperandrogenism in PCOS animal and cell models. These findings contributed the potential therapy by targeting the FTO for hyperandrogenism in PCOS.


Assuntos
Hiperandrogenismo , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Ratos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Androgênios/metabolismo , Di-Hidrotestosterona/metabolismo , Células da Granulosa/metabolismo , Hiperandrogenismo/complicações , Ligases/metabolismo , Síndrome do Ovário Policístico/complicações , Antígeno Prostático Específico/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
Front Endocrinol (Lausanne) ; 14: 1265152, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37929036

RESUMO

Background: Polycystic ovary syndrome (PCOS) is a multifaceted disorder that impacts metabolism, reproduction, as well as endocrine function, characterized by excessive levels of androgen and insulin resistance. The gut microbiota has been implicated in the pathogenesis of PCOS. However, the precise mechanisms through which the gut microbiota influences PCOS still require further elucidation. Methods: The PCOS mouse model was established through the administration of letrozole to both conventional and antibiotics-treated mice. The evaluation of glucose metabolism, sex hormone levels, and ovarian morphology was conducted. Furthermore, the fecal samples from each group of mice were subjected to 16S rRNA gene sequencing, and functional prediction of gut microbiota was proceeded using PICRUSt2 to explore potential mechanisms. Results: By using letrozole-induced PCOS mice model, we manifested that antibiotic intervention significantly reduced the serum total testosterone level and ameliorated glucose intolerance. Antibiotic treatment reduced the number of amplicon sequence variants (ASVs), as well as the Shannon and Simpson index. Meanwhile, letrozole induced a significant increase in the Shannon and Simpson index instead of ASVs. Through random forest model analysis, the results revealed significant alterations in three distinct groups of microbiota, namely Clostridia_vadinBB60_group, Enterorhabdus, and Muribaculaceae after letrozole treatment. Further correlation analysis revealed a positive association between alterations in these microbiota and both serum total testosterone levels and the area under the curve (AUC) of blood glucose in IPGTT. The administration of antibiotics led to a decrease in the absolute abundance of 5 ASVs belonging to unclassified Clostridia_vadinBB60_group, unclassified Enterorhabdus, and unclassified Muribaculaceae, which exhibited a positive correlation with the levels of total testosterone in mice serum, as well as the area under the curve of blood glucose in IPGTT. Moreover, 25 functional pathways of gut microbiome were significantly discrepant between the letrozole-treated mice with and without antibiotics. Conclusion: These results suggest that disturbance of the gut microbiota may take participate in the progression of PCOS and manipulating the composition of the gut microbiota may be a therapeutic approach for managing PCOS.


Assuntos
Microbioma Gastrointestinal , Hiperandrogenismo , Síndrome do Ovário Policístico , Feminino , Humanos , Camundongos , Animais , Síndrome do Ovário Policístico/metabolismo , Letrozol/uso terapêutico , Hiperandrogenismo/complicações , Glicemia/metabolismo , RNA Ribossômico 16S , Testosterona , Antibacterianos/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...