Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 708
Filter
1.
Obesity (Silver Spring) ; 32(4): 678-690, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38439205

ABSTRACT

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.


Subject(s)
Fatty Liver , Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Adolescent , Female , Humans , Hyperandrogenism/complications , Insulin , Lipoproteins, VLDL , Obesity/complications , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/complications
2.
Medicina (Kaunas) ; 60(2)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38399531

ABSTRACT

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.


Subject(s)
Dyslipidemias , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Hyperandrogenism/complications , Hyperandrogenism/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Dyslipidemias/complications , Dyslipidemias/drug therapy
3.
Expert Rev Endocrinol Metab ; 19(2): 111-128, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38205927

ABSTRACT

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.


Subject(s)
Acne Vulgaris , Hyperandrogenism , Adult , Humans , Female , Hirsutism/diagnosis , Hirsutism/etiology , Hyperandrogenism/complications , Hyperandrogenism/diagnosis , Androgens , Dermatologists , Testosterone/metabolism , Alopecia/diagnosis , Alopecia/etiology , Acne Vulgaris/diagnosis , Acne Vulgaris/etiology
4.
Gynecol Obstet Invest ; 89(1): 22-30, 2024.
Article in English | MEDLINE | ID: mdl-38194939

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/epidemiology , Hyperandrogenism/complications , Hyperandrogenism/epidemiology , Quality of Life
5.
Biomol Concepts ; 15(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38242137

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/therapy , Polycystic Ovary Syndrome/complications , Antioxidants/therapeutic use , Hyperandrogenism/complications , Oxidative Stress
6.
Horm Res Paediatr ; 97(2): 134-139, 2024.
Article in English | MEDLINE | ID: mdl-37552972

ABSTRACT

INTRODUCTION: The prevalence of polycystic ovarian syndrome (PCOS) in adolescent girls is between 1 and 4.3%. It remains controversial whether women with a history of idiopathic central precocious puberty (ICPP) are at increased risk for PCOS. Our objective was to assess the prevalence of PCOS in adolescents with a history of ICPP compared with healthy adolescents and the prevalence of PCOS among ICPP girls who have received or not gonadotropin-releasing hormone analogue (GnRHa) treatment. METHODS: We assessed post-menarcheal girls with a history of ICPP. Girls were evaluated at gynecological age ≥2.5 years. Data collected were age at menarche, menstrual cycle characteristics, BMI, clinical hyperandrogenism (HA), total and free testosterone levels. PCOS diagnosis was defined by criteria for adolescents. Subjects were also analyzed regarding whether or not they had received GnRHa treatment. RESULTS: Ninety-four subjects were assessed, and 63 had been treated with GnRHa. Menstrual disorders were found in 29%, clinical HA in 36%, and biochemical HA in 23%. Twelve percent met the diagnostic criteria for PCOS. There was no difference in BMI or in the incidence of menstrual dysfunction or hyperandrogenemia between treated and untreated patients. A higher proportion of clinical HA was found in untreated patients when compared to treated girls. The relative risk (RR) of developing PCOS in ICPP girls was 2.5 compared to a population of healthy adolescents. This RR was not higher in patients who received treatment with GnRHa than in those who did not. CONCLUSION: Adolescent girls with a history of ICPP have an increased risk of PCOS. This risk seems not to be related to GnRHa treatment.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Puberty, Precocious , Adolescent , Female , Humans , Child, Preschool , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/epidemiology , Puberty, Precocious/drug therapy , Prevalence , Hyperandrogenism/complications , Hyperandrogenism/epidemiology , Menarche
7.
J Pediatr Adolesc Gynecol ; 37(1): 51-55, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37696388

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Adolescent , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/diagnosis , Hyperandrogenism/complications , Retrospective Studies , Prevalence , Gender-Affirming Care
8.
Endocrinol Diabetes Metab ; 7(1): e443, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37872876

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Humans , Adult , Female , Mice , Animals , Hyperandrogenism/complications , Hyperandrogenism/metabolism , Dysbiosis/complications , Dysbiosis/metabolism , Diabetes Mellitus, Type 2/complications , RNA, Ribosomal, 16S , Polycystic Ovary Syndrome/complications , Estrogens
9.
J Endocrinol Invest ; 47(4): 927-936, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37907709

ABSTRACT

AIM: To assess which parameters among hyperandrogenism (total testosterone-tT-or free androgen index-FAI), sex hormone binding globulin (SHBG) or body mass index (BMI) could better predict a worse metabolic profile in women with polycystic ovary syndrome (PCOS). METHODS: Five hundred and eighty-six women with PCOS and clinical or biochemical hyperandrogenism were included. Receiver Operating Characteristics (ROC) curves with tT, FAI, SHBG and BMI were performed for metabolic parameters and a cut-off with sensitivity and specificity was obtained for each parameter. The women were then divided into three groups and compared according to their BMI. RESULTS: Based on the ROC curves, tT proved not to be a good predictor of metabolic alterations. FAI and SHBG resulted to be good predictors of some markers of metabolic damage. The area under the curves (AUC) of SHBG were greater than those of FAI. SHBG levels affects the values of homeostasis model assessment of insulin resistance (HOMA-IR), fasting insulin, high density lipoproteins (HDL), low density lipoproteins (LDL), and total cholesterol also when corrected for BMI. However, the highest AUCs of the ROC curves were observed when BMI was used, which was significantly related to all the metabolic parameters analyzed. Dividing women according to their BMI, BMI between 25.00 and 30.00 kg/m2 had a worse metabolic profile but still in a normal range, while BMI ≥ 30 kg/m2 women had a significant metabolic derangement. DISCUSSION: BMI is a good predictor factor of metabolic changes in PCOS women at any age, and obesity is associated to the appearance of metabolic complications. Overweight and obese PCOS women should be addressed to perform a complete metabolic assessment.


Subject(s)
Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Hyperandrogenism/complications , Body Mass Index , Insulin/metabolism , Testosterone , Obesity/metabolism
10.
Article in English | MEDLINE | ID: mdl-37664890

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Diabetes Mellitus, Type 2/etiology , Risk Factors , Hyperandrogenism/complications , Insulin Resistance/physiology
11.
J Pediatr Adolesc Gynecol ; 37(2): 217-219, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38110028

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Leydig Cell Tumor , Ovarian Neoplasms , Male , Child , Humans , Female , Adolescent , Leydig Cell Tumor/complications , Leydig Cell Tumor/surgery , Leydig Cell Tumor/diagnosis , Hyperandrogenism/complications , Virilism/etiology , Ovarian Neoplasms/complications , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/surgery , Androgens
12.
Curr Atheroscler Rep ; 25(12): 1003-1011, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048007

ABSTRACT

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.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Risk Factors , Hyperandrogenism/complications , Hyperandrogenism/diagnosis , Inflammation/complications , Atherosclerosis/complications
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(9): 1425-1431, 2023.
Article in English, Chinese | MEDLINE | ID: mdl-38044655

ABSTRACT

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.


Subject(s)
Acanthosis Nigricans , Hyperandrogenism , Insulin Resistance , Metabolic Syndrome , Metformin , Polycystic Ovary Syndrome , Female , Child , Humans , Hyperandrogenism/complications , Hyperandrogenism/diagnosis , Hyperandrogenism/genetics , Acanthosis Nigricans/complications , Acanthosis Nigricans/diagnosis , Acanthosis Nigricans/genetics , Liraglutide/therapeutic use , Metformin/therapeutic use , Quality of Life , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/complications , Metabolic Syndrome/complications , Metabolic Syndrome/drug therapy
14.
Medicine (Baltimore) ; 102(50): e36075, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38115365

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/drug therapy , Gastrointestinal Microbiome/physiology , Dysbiosis , Hyperandrogenism/complications , Insulin Resistance/genetics
15.
Femina ; 51(12): 682-686, 20231230.
Article in Portuguese | LILACS | ID: biblio-1532471

ABSTRACT

A hiperplasia adrenal congênita (HAC) pode cursar com redução da fertilidade na mulher. Entretanto, nos casos em que ocorre gestação, os recém-nascidos das por- tadoras de hiperplasia adrenal congênita exibem risco de hiperandrogenismo, com todas as suas consequências. A presente revisão atualiza o tema, considerando também as necessidades da assistência a essas pacientes. A busca identificou 294 artigos na base de dados MEDLINE/PubMed de 1961 a março/2023, e os resultados mostraram que as portadoras de hiperplasia adrenal congênita exibem significativa redução da fertilidade. Nos casos de interesse de gestação, as portadoras de hiper- plasia adrenal congênita devem fazer um planejamento reprodutivo, envolvendo a fase antenatal, o acompanhamento pré-natal especializado, o parto e o aleitamento.


Congenital adrenal hyperplasia may lead to reduced male and female fertility. Mo- reover, when the pregnancy occurs, the newborns of patients with congenital adrenal hyperplasia are at risk of hyperandrogenism with all its consequences. This review updates the theme and emphasizes assistance needs. The search identified 294 ar- ticles in the MEDLINE/PubMed database from 1961 to March/2023, and the results showed that patients with congenital adrenal hyperplasia truly exhibit a significant reduction in fertility. In cases of interest in pregnancy, patients with congenital adre- nal hyperplasia should carry out reproductive planning, involving the antenatal pha- se, specialized prenatal care, till delivery and breastfeeding.


Subject(s)
Humans , Male , Female , Adrenal Hyperplasia, Congenital/diagnosis , Reproductive Health , Testicular Neoplasms/complications , Hyperandrogenism/complications , Infertility/complications
16.
Probl Endokrinol (Mosk) ; 69(5): 107-114, 2023 Nov 12.
Article in Russian | MEDLINE | ID: mdl-37968958

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/etiology , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Diabetes Mellitus, Type 2/complications , Hyperandrogenism/complications , Androgens
17.
Front Endocrinol (Lausanne) ; 14: 1265152, 2023.
Article in English | MEDLINE | ID: mdl-37929036

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Mice , Animals , Polycystic Ovary Syndrome/metabolism , Letrozole/therapeutic use , Hyperandrogenism/complications , Blood Glucose/metabolism , RNA, Ribosomal, 16S , Testosterone , Anti-Bacterial Agents/adverse effects
18.
Front Endocrinol (Lausanne) ; 14: 1191759, 2023.
Article in English | MEDLINE | ID: mdl-37929034

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women of reproductive age. Although promising strides have been made in the field of PCOS over the past decades, the distinct etiologies of this syndrome are not fully elucidated. Prenatal factors, genetic variation, epigenetic mechanisms, unhealthy lifestyles, and environmental toxins all contribute to the development of this intricate and highly heterogeneous metabolic, endocrine, reproductive, and psychological disorder. Moreover, interactions between androgen excess, insulin resistance, disruption to the hypothalamic-pituitary-ovary (HPO) axis, and obesity only make for a more complex picture. In this review, we investigate and summarize the related molecular mechanisms underlying PCOS pathogenesis from the perspective of the level of signaling pathways, including PI3K/Akt, TGF-ß/Smads, Wnt/ß-catenin, and Hippo/YAP. Additionally, this review provides an overview of prospective therapies, such as exosome therapy, gene therapy, and drugs based on traditional Chinese medicine (TCM) and natural compounds. By targeting these aberrant pathways, these interventions primarily alleviate inflammation, insulin resistance, androgen excess, and ovarian fibrosis, which are typical symptoms of PCOS. Overall, we hope that this paper will pave the way for better understanding and management of PCOS in the future.


Subject(s)
Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Pregnancy , Female , Humans , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/etiology , Hyperandrogenism/complications , Androgens/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
19.
Gynecol Endocrinol ; 39(1): 2276167, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37931646

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Animals , Female , Humans , Rats , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Androgens/metabolism , Dihydrotestosterone/metabolism , Granulosa Cells/metabolism , Hyperandrogenism/complications , Ligases/metabolism , Polycystic Ovary Syndrome/complications , Prostate-Specific Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism
20.
Obstet Gynecol Clin North Am ; 50(4): 695-705, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37914488

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/therapy , Hyperandrogenism/therapy , Hyperandrogenism/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...