Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.629
Filter
1.
Rev Assoc Med Bras (1992) ; 70(4): e20231368, 2024.
Article in English | MEDLINE | ID: mdl-38716947

ABSTRACT

OBJECTIVE: The study evaluated the opinions of polycystic ovary syndrome on the life quality of women. METHODS: A total of 249 women with polycystic ovary syndrome participated in this descriptive study between October 2022 and July 2023 in Istanbul, Turkey. FINDINGS: Polycystic Ovary Syndrome and Quality of Life was significantly correlated with age (p=0.000) and frequent weight loss diets (p=0.000) (p<0.01). Among the Polycystic Ovary Syndrome and Quality of Life total score and polycystic ovary syndrome symptoms, those with hormone imbalance and insulin resistance had the highest mean scores, while those with menstrual irregularity and fatigue had the lowest. CONCLUSION: Advancing age changes the quality of life of women with polycystic ovary syndrome. To prevent the negative impact of polycystic ovary syndrome on women's quality of life, it is recommended that health professionals develop effective care plans utilizing available evidence.


Subject(s)
Polycystic Ovary Syndrome , Quality of Life , Humans , Polycystic Ovary Syndrome/psychology , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Female , Adult , Young Adult , Turkey , Surveys and Questionnaires , Adolescent , Age Factors , Insulin Resistance/physiology , Body Mass Index
2.
BMC Womens Health ; 24(1): 282, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724955

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is the most prevalent cause of ovulatory infertility and endocrine abnormalities in reproductive-age women. Although the MIND diet has been introduced to improve brain function, evidence shows that the MIND diet is rich in beneficial food groups that can have a preventive effect on other metabolic disorders. The present study was conducted to investigate the association between adherence to the MIND diet and PCOS. METHODS: This age and BMI frequency-matched case-control study was conducted on 216 women between January 2018 and March 2019 in Yazd, Iran. PCOS was diagnosed based on Rotterdam criteria. Participants were selected by convenience sampling method. The validated 178-item food frequency questionnaire was used to assess the usual dietary intake. Logistic regression was used to estimate the association between the MIND diet and PCOS. RESULTS: The findings of the present study showed a significant inverse association between adherence to the MIND diet and PCOS in the crude model (OR for T3 vs. T1: 0.12 (95% CI: 0.05-0.25), P-value < 0.001) and multivariable-adjusted model including energy intake, age, BMI, waist circumference, marital status, pregnancy history, drug use history, education and physical activity (OR for T3 vs. T1 = 0.08 (95% CI: 0.03-0.19), P-value < 0.001). Adherence to the MIND diet had a protective effect of 92%. CONCLUSION: Although the results of the present study showed that higher adherence to the MIND diet is associated with a lower risk of PCOS, more studies are needed to confirm these findings in the future.


Subject(s)
Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/complications , Case-Control Studies , Adult , Iran/epidemiology , Diet/methods , Diet/statistics & numerical data , Young Adult , Body Mass Index
3.
JAMA Netw Open ; 7(5): e249657, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38700861

ABSTRACT

Importance: Polycystic ovary syndrome (PCOS), characterized by irregular menstrual cycles and hyperandrogenism, is a common ovulatory disorder. Having an irregular cycle is a potential marker for cardiometabolic conditions, but data are limited on whether the associations differ by PCOS status or potential interventions. Objective: To evaluate the association of PCOS, time to regularity since menarche (adolescence), and irregular cycles (adulthood) with cardiometabolic conditions. Design, Setting, and Participants: This cross-sectional study used a large, US-based digital cohort of users of the Apple Research application on their iPhone. Eligibility criteria were having ever menstruated, living in the US, being at age of consent of at least 18 years (or 19 years in Alabama and Nebraska or 21 years in Puerto Rico), and being able to communicate in English. Participants were enrolled between November 14, 2019, and December 13, 2022, and completed relevant surveys. Exposures: Self-reported PCOS diagnosis, prolonged time to regularity (not spontaneously establishing regularity within 5 years of menarche), and irregular cycles. Main Outcomes and Measures: The primary outcome was self-reported cardiometabolic conditions, including obesity, prediabetes, type 1 and 2 diabetes, high cholesterol, hypertension, metabolic syndrome, arrhythmia, congestive heart failure, coronary artery disease, heart attack, heart valve disease, stroke, transient ischemic attack (TIA), deep vein thrombosis, and pulmonary embolism measured using descriptive statistics and logistic regression to estimate prevalence odds ratios (PORs) and 95% CIs. Effect modification by lifestyle factors was also estimated. Results: The study sample (N = 60 789) had a mean (SD) age of 34.5 (11.1) years, with 12.3% having PCOS and 26.3% having prolonged time to regularity. Among a subset of 25 399 participants who completed the hormonal symptoms survey, 25.6% reported irregular cycles. In covariate-adjusted logistic regression models, PCOS was associated with a higher prevalence of all metabolic and several cardiovascular conditions, eg, arrhythmia (POR, 1.37; 95% CI, 1.20-1.55), coronary artery disease (POR, 2.92; 95% CI, 1.95-4.29), heart attack (POR, 1.79; 95% CI, 1.23-2.54), and stroke (POR, 1.66; 95% CI, 1.21-2.24). Among participants without PCOS, prolonged time to regularity was associated with type 2 diabetes (POR, 1.24; 95% CI, 1.05-1.46), hypertension (POR, 1.09; 95% CI, 1.01-1.19), arrhythmia (POR, 1.20; 95% CI, 1.06-1.35), and TIA (POR, 1.33; 95% CI, 1.01-1.73), and having irregular cycles was associated with type 2 diabetes (POR, 1.36; 95% CI, 1.08-1.69), high cholesterol (POR, 1.17; 95% CI, 1.05-1.30), arrhythmia (POR, 1.21; 95% CI, 1.02-1.43), and TIA (POR, 1.56; 95% CI, 1.06-2.26). Some of these associations were modified by high vs low body mass index or low vs high physical activity. Conclusions and Relevance: These findings suggest that PCOS and irregular cycles may be independent markers for cardiometabolic conditions. Early screening and intervention among individuals with irregular menstrual cycles may be beneficial.


Subject(s)
Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/complications , Cross-Sectional Studies , Adult , Menstruation Disturbances/epidemiology , United States/epidemiology , Cardiovascular Diseases/epidemiology , Young Adult , Cohort Studies , Middle Aged , Obesity/epidemiology , Adolescent , Alabama/epidemiology
4.
Endocr Regul ; 58(1): 83-90, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38656255

ABSTRACT

Objective. Abnormal lipid profile and obesity increase the risk of polycystic ovary syndrome (PCOS). PCOS patients may have a greater risk of infertility, metabolic syndrome (MetS) and cardiovascular disease (CVD) due to abnormal lipid profile and obesity. The aim of the study was to find the association between abnormal lipid profile and obesity in patients with PCOS. Methods. In this case-control study, a total of 102 female subjects (51 diagnosed PCOS and 51 age-matched healthy controls) were enrolled, aged between 20-40 years. Biochemical parameters such as total cholesterol (TC), triglycerides (TG), low-density lipoprotein-cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), very low-density lipoprotein-cholesterol (VLDL-C), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) were estimated. Anthropometric parameters such as body mass index (BMI), waist circumference (WC), hip circumference (HC), and waist-to-hip ratio (WHR) were recorded. A p<0.05 was considered statistically significant. Results. Mean of BMI, WC, WHR, LH, FSH, TC, TG, LDL-C, and VLDL-C was found significantly elevated in patients with PCOS as compared to controls (p<0.01). However, the mean of HDL-C was found significantly reduced in patients with PCOS as compared to controls (p<0.01). BMI has shown a significant positive correlation with WC (r=0.562, p<0.01) and WHR (r=0.580, p<0.01) among PCOS patients. LH has shown a significant positive correlation with FSH (r=0.572, p<0.01) among PCOS patients. TC has shown a significant positive correlation with TG (r=0.687, p<0.01), LDL-C (r=0.917, p<0.01), and VLDL-C (r=0.726, p<0.01) among PCOS patients. Conclusion. The results showed that abnormal lipid profile and obesity have a significant association with PCOS patients. Regular monitoring and treatment of PCOS patients are required to reduce the risk of infertility, MetS, and CVD.


Subject(s)
Body Mass Index , Lipids , Obesity , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Female , Adult , Case-Control Studies , Young Adult , Obesity/blood , Obesity/complications , Lipids/blood , Waist Circumference , Triglycerides/blood , Luteinizing Hormone/blood , Waist-Hip Ratio , Follicle Stimulating Hormone/blood , Cholesterol, LDL/blood
5.
BMJ Open ; 14(4): e070801, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684265

ABSTRACT

INTRODUCTION: Low vitamin D status is prevalent among women with polycystic ovary syndrome (PCOS). The objective of the study is to assess the effect of vitamin D supplementation on (1) the ovulation rate to letrozole and (2) other reproductive, endocrine and metabolic outcomes after 1 year of supplementation in women with PCOS. METHODS AND ANALYSIS: This is a multicentre, randomised, double-blind, controlled clinical trial. A total of 220 anovulatory women with PCOS diagnosed by the Rotterdam criteria will be recruited. They will be randomly assigned to either the (1) vitamin D supplementation group or (2) placebo group. Those in the vitamin D group will take oral Vitamin D3 50 000 IU/week for 4 weeks, followed by 50 000 IU once every 2 weeks for 52 weeks. Those who remain anovulatory after 6 months will be treated with a 6-month course of letrozole (2.5 mg to 7.5 mg for 5 days per cycle titrated according to response) for ovulation induction. The primary outcome is the ovulation rate. All statistical analyses will be performed using intention-to-treat and per protocol analyses. ETHICS AND DISSEMINATION: Ethics approval was sought from the Institutional Review Board of the participating units. All participants will provide written informed consent before joining the study. The results of the study will be submitted to scientific conferences and peer-reviewed journals. TRIAL REGISTRATION NUMBER: NCT04650880.


Subject(s)
Letrozole , Ovulation Induction , Ovulation , Polycystic Ovary Syndrome , Adult , Female , Humans , Young Adult , Aromatase Inhibitors/therapeutic use , Aromatase Inhibitors/administration & dosage , Dietary Supplements , Double-Blind Method , Letrozole/therapeutic use , Letrozole/administration & dosage , Multicenter Studies as Topic , Ovulation/drug effects , Ovulation Induction/methods , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/complications , Randomized Controlled Trials as Topic , Vitamin D/therapeutic use , Vitamin D/administration & dosage
6.
BMC Womens Health ; 24(1): 234, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38610020

ABSTRACT

BACKGROUND: People with polycystic ovary syndrome suffer from many symptoms and are at risk of developing diseases such as hypertension and diabetes in the future. Therefore, the importance of self-care doubles. It is mainly to modify the lifestyle, especially following the principles of healthy eating. The purpose of this study is to review artificial intelligence-based systems for providing management recommendations, especially food recommendations. MATERIALS AND METHODS: This study started by searching three databases: PubMed, Scopus, and Web of Science, from inception until 6 June 2023. The result was the retrieval of 15,064 articles. First, we removed duplicate studies. After the title and abstract screening, 119 articles remained. Finally, after reviewing the full text of the articles and considering the inclusion and exclusion criteria, 20 studies were selected for the study. To assess the quality of articles, we used criteria proposed by Malhotra, Wen, and Kitchenham. Out of the total number of included studies, seventeen studies were high quality, while three studies were moderate quality. RESULTS: Most studies were conducted in India in 2021. Out of all the studies, diagnostic recommendation systems were the most frequently researched, accounting for 86% of the total. Precision, sensitivity, specificity, and accuracy were more common than other performance metrics. The most significant challenge or limitation encountered in these studies was the small sample size. CONCLUSION: Recommender systems based on artificial intelligence can help in fields such as prediction, diagnosis, and management of polycystic ovary syndrome. Therefore, since there are no nutritional recommendation systems for these patients in Iran, this study can serve as a starting point for such research.


Subject(s)
Hypertension , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/therapy , Artificial Intelligence , Diet, Healthy , India
7.
Nutrients ; 16(7)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38613082

ABSTRACT

Polycystic ovary syndrome (PCOS), the most common endocrine disorder in women of reproductive age, constitutes a metabolic disorder frequently associated with obesity and insulin resistance (IR). Furthermore, women with PCOS often suffer from excessive anxiety and depression, elicited by low self-esteem due to obesity, acne, and hirsutism. These mood disorders are commonly associated with food cravings and binge eating. Hypothalamic signaling regulates appetite and satiety, deteriorating excessive food consumption. However, the hypothalamic function is incapable of compensating for surplus food in women with PCOS, leading to the aggravation of obesity and a vicious circle. Hyperandrogenism, IR, the reduced secretion of cholecystokinin postprandially, and leptin resistance defined by leptin receptors' knockout in the hypothalamus have been implicated in the pathogenesis of hypothalamic dysfunction and appetite dysregulation. Diet modifications, exercise, and psychological and medical interventions have been applied to alleviate food disorders, interrupting the vicious circle. Cognitive-behavioral intervention seems to be the mainstay of treatment, while the role of medical agents, such as GLP-1 analogs and naltrexone/bupropion, has emerged.


Subject(s)
Acne Vulgaris , Polycystic Ovary Syndrome , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/therapy , Craving , Obesity/complications , Obesity/therapy , Appetite
8.
Rev Assoc Med Bras (1992) ; 70(3): e20230918, 2024.
Article in English | MEDLINE | ID: mdl-38655994

ABSTRACT

OBJECTIVE: The objective of this study was to learn more about the prevalence and pathophysiology of depression and anxiety that may be caused by polycystic ovary syndrome and to make plans for taking necessary precautions for this vulnerable group. METHODS: This case-control study was conducted between January 2022 and October 2022. A total of 120 women with polycystic ovary syndrome and 143 controls were included in the study. All healthy volunteers and women with polycystic ovary syndrome were evaluated using self-administered questionnaires and physical examination. Anthropometric data such as weight and height and laboratory value were documented. RESULTS: There was no significant difference between the groups in terms of demographic characteristics. When the Hospital Anxiety and Depression Scale scores of both groups were compared, both depression and anxiety scores were found to be significantly higher in women with polycystic ovary syndrome compared with the control group (OR: 3.319, 95%CI, 1.563-7.047, p<0.001 and OR: 3.238, 95%CI, 1.659-6.315, p<0.001). In the Hospital Anxiety and Depression Scale questionnaire, the rate of irregular menstruation and Ferriman-Gallwey score were statistically significant in women with polycystic ovary syndrome with high depression and anxiety scores. While serum LH levels and LH/FSH ratios were significantly different in women with polycystic ovary syndrome with high depression scores, serum LH, LH:FSH ratios, and serum total testosterone levels were found significant in women with polycystic ovary syndrome with high anxiety scores. CONCLUSION: It is clear that depression and anxiety are more common in patients with polycystic ovary syndrome than in healthy women. Our findings support previous recommendations regarding routine screening for depression and anxiety in this population.


Subject(s)
Anxiety , Depression , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/psychology , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/blood , Female , Cross-Sectional Studies , Case-Control Studies , Adult , Risk Factors , Depression/epidemiology , Depression/etiology , Anxiety/etiology , Anxiety/epidemiology , Young Adult , Surveys and Questionnaires , Prevalence , Adolescent , Psychiatric Status Rating Scales , Luteinizing Hormone/blood
9.
Reprod Biol Endocrinol ; 22(1): 43, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627777

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder associated with infertility and pregnancy complications. The pathogenesis of PCOS and its impact on reproductive function may be influenced by the source of androgens, including testosterone, free androgen, dehydroepiandrosterone sulfate (DHEAS). However, the differential effects of these androgen on pregnancy and neonatal outcomes and the cut-off value of East Asian population with PCOS remain unclear. METHODS: A retrospective cohort study was conducted at the Reproductive Medicine Center of the First Affiliated Hospital of Sun Yat-sen University from January 2015 to November 2022, involving 636 cycles of in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI). Subgroup analyses were performed using cut-off values of 6.4 for free androgen index (FAI), 9.5 µmol/L for DHEAS. Pregnancy and neonatal outcomes were compared between groups. Restricted cubic spline (RCS) was used to identify significant cut-off values affecting pregnancy. RESULTS: Higher FAI levels (> 6.4) were associated with decrease in clinical pregnancy rate (PR) (50.61% vs. 41.66%, p = 0.024), live birth rate (LBR) (42.42% vs. 32.35%, p = 0.011). When DHEAS levels exceeded 9.5 µmol/L, there was a significant decrease in clinical PR (51.27% vs. 42.73%, P = 0.039), LBR (42.73% vs. 32.73%, P = 0.012). Negative correlations were also observed between DHEAS levels and cumulative pregnancy rate (70.57% vs 56.62% p = 0.002) and cumulative live birth rate (CLBR) (59.35% vs 43.37%, p = 0.0007). Both FAI and DHEAS elevated is associated with the lowest clinical pregnancy rate (37.84%). Conversely, when solely FAI is elevated, the pregnancy rate increases to 52.38%, while an elevation in DHEAS alone is associated with a pregnancy rate of, both of which are lower than when neither FAI nor DHEAS are elevated (60.68%). The live birth rates exhibit a similar trend (30.00% vs 40.00% vs 41.83% vs 44.48%). RCS revealed a significant decrease in CPR and CLBR when DHEA levels exceeded 7.69 umol/L, while the cut-off value of FAI was 6.36 for CPR and CLBR. CONCLUSION: In conclusion, PCOS patients with biochemical hyperandrogenism show unsatisfactory clinical PR and CLBR when undergoing assisted reproductive technology (ART). This may be attributed to the influence of both adrenal-derived DHEAS and ovarian-derived FAI on the unfavorable pregnancy outcomes.


Subject(s)
Polycystic Ovary Syndrome , Male , Pregnancy , Female , Infant, Newborn , Humans , Polycystic Ovary Syndrome/complications , Androgens , Dehydroepiandrosterone Sulfate , Retrospective Studies , Semen , Dehydroepiandrosterone
10.
Gynecol Endocrinol ; 40(1): 2341701, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38622970

ABSTRACT

OBJECTIVE: To evaluate the effects of alpha lipoic acid (ALA) on hormonal and metabolic parameters in a group of overweight/obese Polycystic Ovary Syndrome (PCOS) patients. METHODS: This was a retrospective study in which thirty-two overweight/obese patients with PCOS (n = 32) not requiring hormonal treatment were selected from the database of the ambulatory clinic of the Gynecological Endocrinology Center at the University of Modena and Reggio Emilia, Italy. The hormonal profile, routine exams and insulin and C-peptide response to oral glucose tolerance test (OGTT) were evaluated before and after 12 weeks of complementary treatment with ALA (400 mg/day). Hepatic Insulin Extraction (HIE) index was also calculated. RESULTS: ALA administration significantly improved insulin sensitivity and decreased ALT and AST plasma levels in all subjects, though no changes were observed on reproductive hormones. When PCOS patients were subdivided according to the presence or absence of familial diabetes background, the higher effects of ALA were observed in the former group that showed AST and ALT reduction and greater HIE index decrease. CONCLUSION: ALA administration improved insulin sensitivity in overweight/obese PCOS patients, especially in those with familial predisposition to diabetes. ALA administration improved both peripheral sensitivity to insulin and liver clearance of insulin. Such effects potentially decrease the risk of nonalcoholic fat liver disease and diabetes in PCOS patients.


Subject(s)
Diabetes Mellitus , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Polycystic Ovary Syndrome , Thioctic Acid , Female , Humans , Insulin , Insulin Resistance/physiology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Obesity/drug therapy , Overweight/complications , Overweight/drug therapy , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Retrospective Studies , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use
12.
Nat Rev Dis Primers ; 10(1): 27, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637590

ABSTRACT

Despite affecting ~11-13% of women globally, polycystic ovary syndrome (PCOS) is a substantially understudied condition. PCOS, possibly extending to men's health, imposes a considerable health and economic burden worldwide. Diagnosis in adults follows the International Evidence-based Guideline for the Assessment and Management of Polycystic Ovary Syndrome, requiring two out of three criteria - clinical or biochemical hyperandrogenism, ovulatory dysfunction, and/or specific ovarian morphological characteristics or elevated anti-Müllerian hormone. However, diagnosing adolescents omits ovarian morphology and anti-Müllerian hormone considerations. PCOS, marked by insulin resistance and hyperandrogenism, strongly contributes to early-onset type 2 diabetes, with increased odds for cardiovascular diseases. Reproduction-related implications include irregular menstrual cycles, anovulatory infertility, heightened risks of pregnancy complications and endometrial cancer. Beyond physiological manifestations, PCOS is associated with anxiety, depression, eating disorders, psychosexual dysfunction and negative body image, collectively contributing to diminished health-related quality of life in patients. Despite its high prevalence persisting into menopause, diagnosing PCOS often involves extended timelines and multiple health-care visits. Treatment remains ad hoc owing to limited understanding of underlying mechanisms, highlighting the need for research delineating the aetiology and pathophysiology of the syndrome. Identifying factors contributing to PCOS will pave the way for personalized medicine approaches. Additionally, exploring novel biomarkers, refining diagnostic criteria and advancing treatment modalities will be crucial in enhancing the precision and efficacy of interventions that will positively impact the lives of patients.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperandrogenism , Polycystic Ovary Syndrome , Pregnancy , Adult , Adolescent , Female , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/diagnosis , Diabetes Mellitus, Type 2/complications , Quality of Life , Anti-Mullerian Hormone
13.
Med Sci Monit ; 30: e943594, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38600677

ABSTRACT

BACKGROUND The endocrine metabolic disorder polycystic ovary syndrome (PCOS) is quite common among women. Women with PCOS show a compromised health-related quality of life (HRQoL), impaired emotional well-being, and alexithymia, a neuropsychological phenomenon conceptualized as difficulty in recognizing and expressing one's emotions. However, the relationship between alexithymia and HRQoL in women with PCOS is unclear. This study aimed to investigate the impact of alexithymia on HRQoL in women with PCOS in Saudi Arabia. MATERIAL AND METHODS This descriptive cross-sectional, community-based study enrolled 518 female patients with a diagnosis of PCOS from all age groups. The patients were surveyed via a self-administered questionnaire from December 2022 to January 2023. The survey encompassed sociodemographic data, the 20-item Toronto Alexithymia Scale (TAS-20), and HRQoL 15-dimensional scale (15D). RESULTS This study included 320 participants (61.8%) with alexithymia. A significant association was found between alexithymia and low HRQoL 15D scores in patients with PCOS. Specifically, scores on the TAS-20 subscales related to difficulty in identifying feelings and describing feelings were positively correlated with HRQoL (P=0.000). However, externally oriented thinking subscale scores were not positively correlated with HRQoL (P=0.44). CONCLUSIONS The presence of alexithymia is associated with poor HRQoL in women with PCOS, which is more pronounced among those who received a diagnosis ≥4 years ago. Alexithymia must be considered when assessing the HRQoL of patients with PCOS, considering the relatively high prevalence of alexithymia in these patients.


Subject(s)
Polycystic Ovary Syndrome , Quality of Life , Humans , Female , Quality of Life/psychology , Affective Symptoms/diagnosis , Affective Symptoms/epidemiology , Affective Symptoms/psychology , Polycystic Ovary Syndrome/complications , Cross-Sectional Studies , Saudi Arabia
14.
Gynecol Endocrinol ; 40(1): 2330655, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38613449

ABSTRACT

OBJECTIVE: This study aims to examine the short-term effects of oral metformin (MET) on serum anti-müllerian hormone (AMH) levels and to verify its impact on AMH concentrations in women with polycystic ovary syndrome (PCOS). METHODS: The literature search, extending from January 2000 to April 2023, was conducted using databases such as PubMed, Embase, and the Cochrane Central, resulting in the inclusion of 20 studies. These selected studies, evaluated for quality using the Newcastle-Ottawa Scale, investigated changes in AMH levels before and after treatment, with durations ranging from less than three months to over six months. The reported outcomes were quantified as standardized mean differences (SMD) with 95% confidence intervals (CI). This comprehensive systematic review and meta-analysis was registered with the International Prospective Register of Systematic Reviews (PROSPERO) under the registration number CRD42023420705. The statistical analyses were performed using Review Manager 5.4.1. RESULTS: ① The study incorporated 20 articles, consisting of 12 prospective studies, 7 randomized controlled trials (RCT), and 1 cross-sectional study. ② Serum AMH levels in patients with PCOS diminish subsequent to the oral administration of MET. ③ Across the spectrum of studies analyzed, a pronounced degree of heterogeneity is evident, potentially ascribed to differential parameters including body mass index (BMI), daily pharmacological dosages, the temporal extent of treatment regimens, criteria of PCOS, and detection Methods. ④ The impact of MET on AMH levels exhibits a dose-responsive trend, with escalating doses of MET being associated with progressively greater declines in AMH concentrations in the patient population. ⑤ For women with PCOS receiving MET therapy, a minimum treatment duration of three months may be necessary to observe a reduction in serum AMH levels. CONCLUSIONS: The results of this meta-analysis indicate that MET treatment exerts a suppressive effect on serum AMH levels in women with PCOS. It appears that a treatment duration of at least three months is required to achieve a significant decrease in AMH concentrations. Furthermore, the influence of MET on AMH is dose-dependent, with higher doses correlating with more pronounced reductions in AMH levels among the patients studied.


Subject(s)
Metformin , Peptide Hormones , Polycystic Ovary Syndrome , Female , Humans , Anti-Mullerian Hormone , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Administration, Oral , Body Mass Index , Metformin/therapeutic use
15.
Pediatrics ; 153(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38634159

ABSTRACT

OBJECTIVE: Polycystic Ovary Syndrome (PCOS) is common among females, with significant metabolic and reproductive comorbidities. We describe PCOS development in a pediatric population. METHODS: We assessed cardiometabolic biomarkers and adiposity at the midchildhood (mean 7.9 y), early teen (mean 13.1 y), and midteen (mean 17.8 y) visits among 417 females in the prospective Project Viva cohort. We defined PCOS via self-reported diagnosis or ovulatory dysfunction with hyperandrogenism in midlate adolescence. We used multivariable logistic regression to assess associations of metabolic and adiposity markers at each visit with PCOS. RESULTS: Adolescents with PCOS (n = 56, 13%) versus without had higher mean (SD) BMI z-score and truncal fat mass at the midchildhood (0.66 [0.99] vs 0.30 [1.04]; 3.5 kg [2.6] vs 2.7 [1.5]), early teen (0.88 [1.01] vs 0.25 [1.08]; 9.4 kg [6.7] vs 6.1 [3.4]), and midteen (0.78 [1.03] vs 0.33 [0.97]; 11.6 kg [7.2] vs 9.1 [4.9]) visits as well as lower adiponectin to leptin ratio at the early (0.65 [0.69] vs 1.04 [0.97]) and midteen (0.33 [0.26] vs 0.75 [1.21]) visits. In models adjusted for maternal PCOS, education and child race and ethnicity (social factors), we found higher odds of PCOS per 1-SD increase in truncal fat at midchildhood (odds ratio [OR] 1.42; 95% confidence interval [CI] 1.03-1.95) and early teen visits (OR 1.61; 95% CI 1.14-2.28) and lower odds per 1-SD increase in adiponectin/leptin ratio at the midteen visit (OR 0.14; 95% CI 0.03-0.58). CONCLUSIONS: Childhood excess adiposity and adipose tissue dysfunction may be a first signs of later PCOS risk.


Subject(s)
Adiposity , Biomarkers , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/complications , Female , Adolescent , Child , Biomarkers/blood , Prospective Studies , Adiponectin/blood , Leptin/blood , Body Mass Index
16.
BMC Womens Health ; 24(1): 221, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580996

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) has previously been associated with several comorbidities that may have shared genetic, epigenetic, developmental or environmental origins. PCOS may be influenced by prenatal androgen excess, poor intrauterine or childhood environmental factors, childhood obesity and learned health risk behaviors. We analyzed the association between PCOS and several relevant comorbidities while adjusting for early-life biological and socioeconomic conditions, also investigating the extent to which the association is affected by familial risk factors. METHODS: This total-population register-based cohort study included 333,999 full sisters, born between 1962 and 1980. PCOS and comorbidity diagnoses were measured at age 17-45 years through national hospital register data from 1997 to 2011, and complemented with information on the study subjects´ early-life and social characteristics. In the main analysis, sister fixed effects (FE) models were used to control for all time-invariant factors that are shared among sisters, thereby testing whether the association between PCOS and examined comorbidities is influenced by unobserved familial environmental, social or genetic factors. RESULTS: Three thousand five hundred seventy women in the Sister sample were diagnosed with PCOS, of whom 14% had obesity, 8% had depression, 7% had anxiety and 4% experienced sleeping, sexual and eating disorders (SSE). Having PCOS increased the odds of obesity nearly 6-fold (adjusted OR (aOR): 5.9 [95% CI:5.4-6.5]). This association was attenuated in models accounting for unobserved characteristics shared between full sisters, but remained considerable in size (Sister FE: aOR: 4.5 [95% CI: 3.6-5.6]). For depression (Sister FE: aOR: 1.4 [95% CI: 1.2-1.8]) and anxiety (Sister FE: aOR: 1.5 [95% CI: 1.2-1.8), there was a small decrease in the aORs when controlling for factors shared between sisters. Being diagnosed with SSE disorders yielded a 2.4 aOR (95% CI:2.0-2.6) when controlling for a comprehensive set of individual-level confounders, which only decreased slightly when controlling for factors at the family level such as shared genes or parenting style. Accounting for differences between sisters in observed early-life circumstances influenced the estimated associations marginally. CONCLUSION: Having been diagnosed with PCOS is associated with a markedly increased risk of obesity and sleeping, sexual and eating disorders, also after accounting for factors shared between sisters and early-life conditions.


Subject(s)
Pediatric Obesity , Polycystic Ovary Syndrome , Child , Pregnancy , Female , Humans , Adolescent , Young Adult , Adult , Middle Aged , Polycystic Ovary Syndrome/complications , Cohort Studies , Siblings , Pediatric Obesity/complications , Comorbidity
17.
Front Endocrinol (Lausanne) ; 15: 1366940, 2024.
Article in English | MEDLINE | ID: mdl-38665260

ABSTRACT

Background: Withdrawal of semaglutide is frequently followed by weight regain due to compensatory biological changes that prevent the maintenance of long-term weight loss. There are some studies implying that metformin might attenuate weight regain. The weight trajectory after discontinuation of short-term semaglutide treatment in obese women with PCOS who continued metformin treatment has not yet been evaluated. Aims: We explored changes in body weight, cardiometabolic and endocrine parameters in obese women with PCOS who continued treatment with metformin 2 years after discontinuation of short-term intervention with semaglutide. Methods: 25 women with PCOS and obesity, aged 33.7 ± 5.3 years (mean ± SD), were treated with once-weekly subcutaneous semaglutide 1.0 mg as an adjunct to metformin 2000 mg/day and lifestyle intervention for 16 weeks. At week 16, semaglutide was discontinued. Treatment with metformin 2000 mg/day and promotion of lifestyle intervention were continued during the 2-year follow-up period. Weight change, cardiometabolic, and endocrine parameters were assessed 2 years after semaglutide discontinuation. Results: During semaglutide treatment phase, weight decreased from 101 (90-106.8) kg to 92 (83.3-100.8) kg. Two years after semaglutide withdrawal, weight was 95 (77-104) kg. The net weight loss 2 years after discontinuation of semaglutide remained significant when compared to baseline (p=0.003). At the end of the study, 21 out of 25 subjects had lower body weight compared to baseline. Improvements in cardiometabolic parameters including decrease in total and LDL cholesterol, fasting glucose, and glucose after OGTT that had been seen during semaglutide-treatment phase reverted towards baseline two years after semaglutide cessation. Free testosterone levels significantly decreased during semaglutide treatment from 6.16 (4.07-9.71) to 4.12 (2.98-6.93) nmol/l, (p= 0.012) and did not significantly deteriorate after semaglutide discontinuation. Conclusion: Two years after semaglutide withdrawal, women with PCOS who continued with metformin regained about one-third of the semaglutide-induced weight loss. At the end of the follow up, 84% of women had a lower body weight than at baseline.


Subject(s)
Glucagon-Like Peptides , Hypoglycemic Agents , Metformin , Obesity , Polycystic Ovary Syndrome , Weight Loss , Humans , Female , Metformin/therapeutic use , Metformin/administration & dosage , Adult , Glucagon-Like Peptides/therapeutic use , Glucagon-Like Peptides/administration & dosage , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/complications , Weight Loss/drug effects , Obesity/drug therapy , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/administration & dosage , Follow-Up Studies
18.
Obes Rev ; 25(6): e13737, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491863

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common endocrine disorder in females. Modest weight loss improves reproductive and metabolic PCOS features. While lifestyle modifications and pharmacotherapies remain first-line weight loss strategies, bariatric surgery is emerging as a potentially effective treatment. We performed a systematic review and meta-analysis of published literature to examine the impact of bariatric surgery in PCOS to inform the 2023 International PCOS Evidence-based Guidelines. Electronic databases were searched for observational studies and trials comparing pharmacologic or lifestyle treatments to bariatric surgery in women with PCOS or bariatric surgery in women with or without PCOS. Anthropometric, reproductive, hormonal, and metabolic outcomes were included and, where possible, meta-analyzed using random-effects models. Risk of bias and evidence quality were assessed. Ten studies were included involving 432 women with and 590 women without PCOS. Comparisons between bariatric surgery and pharmacologic or lifestyle treatments were only reported in one study each, and most reproductive outcomes were limited to a single study; therefore, meta-analyses could not be performed. Meta-analysis found that women with PCOS experience similar improvements in anthropometric, hormonal, and metabolic outcomes after bariatric surgery compared to those without PCOS. Existing research is limited and of low quality with high risk of bias, especially in comparison to existing PCOS treatments and with respect to reproductive outcomes including pregnancy, highlighting the need for additional studies to inform clinical recommendations.


Subject(s)
Bariatric Surgery , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/therapy , Female , Treatment Outcome , Pregnancy , Obesity/surgery , Obesity/complications , Weight Loss/physiology
19.
Hum Reprod ; 39(5): 1078-1088, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38503490

ABSTRACT

STUDY QUESTION: Is resumption of ovulation after a 6-month lifestyle intervention in women with PCOS and obesity associated with differential changes in endocrine and metabolic parameters (weight, insulin resistance, anti-Müllerian hormone (AMH), and androgens) compared to women with PCOS who remained anovulatory? SUMMARY ANSWER: Resumption of ovulation after a 6-month lifestyle intervention in women with PCOS and obesity is associated with changes in serum 11ß-hydroxyandrostenedione (11OHA4) concentrations. WHAT IS KNOWN ALREADY: Lifestyle interventions have been shown to reduce clinical and biochemical hyperandrogenism in women with PCOS. Weight loss of 5-10% may reverse anovulatory status, thereby increasing natural conception rates. However, the mechanisms underlying why some women with PCOS remain anovulatory and others resume ovulation after weight loss are unclear. Reproductive characteristics at baseline and a greater degree of change in endocrine and metabolic features with lifestyle intervention may be crucial for ovulatory response. STUDY DESIGN, SIZE, DURATION: We used data and samples originating from an earlier randomized controlled trial (RCT), which examined the efficacy of a 6-month lifestyle intervention prior to infertility treatment compared to prompt infertility treatment on live birth rate in women with obesity. A total of 577 women with obesity (BMI > 29 kg/m2) were randomized between 2009 and 2012. Anovulatory women with PCOS who were allocated to the intervention arm of the original RCT (n = 95) were included in the current analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: We defined women as having resumed ovulation (RO+) based on the following criteria: spontaneous pregnancy; or assignment to expectant management; or IUI in natural cycles as the treatment strategy after lifestyle intervention. Steroid hormones were measured using liquid chromatography tandem mass spectrometry. Generalized estimating equations with adjustment for baseline measures and interaction between group and time was used to examine differences in changes of endocrine and metabolic parameters between RO+ (n = 34) and persistently anovulatory women (RO-, n = 61) at 3 and 6 months after intervention. MAIN RESULTS AND THE ROLE OF CHANCE: At baseline, the mean ± SD age was 27.5 ± 3.6 years in the RO+ group and 27.9 ± 4.1 years in the RO- group (P = 0.65), and the mean ± SD weights were 101.2 ± 9.5 kg and 105.0 ± 14.6 kg, respectively (P = 0.13). Baseline AMH concentrations showed significant differences between RO+ and RO- women (median and interquartile range [IQR] 4.7 [3.2; 8.3] versus 7.2 [5.3; 10.8] ng/ml, respectively). Baseline androgen concentrations did not differ between the two groups. During and after lifestyle intervention, both groups showed weight loss; changes in 11OHA4 were significantly different between the RO+ and RO groups (P-value for interaction = 0.03). There was a similar trend for SHBG (interaction P-value = 0.07), and DHEA-S (interaction P-value = 0.06), with the most pronounced differences observed in the first 3 months. Other parameters, such as AMH and FAI, decreased over time but with no difference between the groups. LIMITATIONS, REASONS FOR CAUTION: No high-resolution transvaginal ultrasonography was used to confirm ovulatory status at the end of the lifestyle program. The small sample size may limit the robustness of the results. WIDER IMPLICATIONS OF THE FINDINGS: Reduction of androgen concentrations during and after lifestyle intervention is associated with recovery of ovulatory cycles. If our results are confirmed in other studies, androgen concentrations could be monitored during lifestyle intervention to provide individualized recommendations on the timing of resumption of ovulation in anovulatory women with PCOS and obesity. STUDY FUNDING/COMPETING INTEREST(S): The study was supported by a grant from ZonMw, the Dutch Organization for Health Research and Development (50-50110-96-518). The Department of Obstetrics and Gynecology of the UMCG received an unrestricted educational grant from Ferring Pharmaceuticals BV, The Netherlands. A.H. reports consultancy for the development and implementation of a lifestyle App MyFertiCoach developed by Ferring Pharmaceutical Company. All other authors have no conflicts to declare. TRIAL REGISTRATION NUMBER: The LIFEstyle RCT was registered at the Dutch trial registry (NTR 1530).


Subject(s)
Anovulation , Obesity , Ovulation , Polycystic Ovary Syndrome , Humans , Female , Obesity/complications , Obesity/therapy , Adult , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/therapy , Androstenedione/blood , Insulin Resistance , Pregnancy , Anti-Mullerian Hormone/blood , Weight Loss
20.
Hum Reprod ; 39(5): 1072-1077, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38514450

ABSTRACT

STUDY QUESTION: What are the characteristics of adolescents diagnosed with polycystic ovary syndrome (PCOS) based on the 2003 Rotterdam criteria, but who do not meet the diagnosis according to the international evidence-based guideline? SUMMARY ANSWER: Adolescents who had features of PCOS but did not meet the evidence-based guideline adolescent criteria exhibited unfavorable metabolic profiles compared to controls and shared considerable metabolic and hormonal features with adolescents who did meet the adolescent criteria. WHAT IS KNOWN ALREADY: The international evidence-based PCOS guideline recommended that ultrasound should not be used for the diagnosis of PCOS in girls with a gynecological age of <8 years. Thus far, few studies have evaluated the clinical characteristics of the girls diagnosed with PCOS based on the Rotterdam criteria but who do not meet the diagnosis according to the updated guideline. STUDY DESIGN, SIZE, DURATION: This is a retrospective study, and subjects attended for care from 2004 to 2022. PARTICIPANTS/MATERIALS, SETTING, METHODS: Adolescent girls with PCOS diagnosed according to the 2003 Rotterdam criteria and healthy controls. All participants were between 2 and 8 years since menarche. MAIN RESULTS AND THE ROLE OF CHANCE: Of the 315 girls diagnosed with PCOS according to the Rotterdam criteria, those with irregular menstruation (IM)/hyperandrogenism (HA)/polycystic ovary (PCO), IM/HA, HA/PCO, and IM/PCO phenotypes accounted for 206 (65.4%), 30 (9.5%), 12 (3.8%), and 67 (21.3%) participants, respectively. According to the evidence-based guideline, 79 girls (25.1%) with the HA/PCO or IM/PCO phenotypes were not diagnosed with PCOS, and aligned to the international guideline; they were designated as the 'at-risk' group. As expected, the girls meeting the evidence-based guideline adolescent criteria showed the worst metabolic profiles (degree of generalized or central obesity, frequency of insulin resistance, prediabetes or diabetes, and metabolic syndrome) and higher hirsutism scores than the at-risk group or controls. Approximately 90% of the at-risk group were not overweight or obese, which was similar to the controls. However, they showed worse metabolic profiles, with higher blood pressure, triglyceride, and insulin resistance parameters than controls; furthermore, these profiles were similar to those of the girls meeting the adolescent criteria. The at-risk group showed similarly elevated serum LH levels and LH/FSH ratio with the girls meeting adolescent criteria. LIMITATIONS, REASONS FOR CAUTION: We could not evaluate hormonal or ultrasound parameters in controls. WIDER IMPLICATIONS OF THE FINDINGS: Compared to the conventional Rotterdam criteria, the recent international evidence-based guideline-avoiding ultrasound in PCOS diagnosis in adolescents-still gives the opportunity to identify young girls at risk, aligned to the findings in this study. A practical approach to this adolescent population would involve establishing IM or HA (with ultrasound not indicated) and designating 'at-risk' PCOS status with regular check-ups for newly developed or worsening PCOS-related symptoms or metabolic abnormalities, with subsequent reassessment including ultrasound or anti-Müllerian hormone, once 8 years post-menarche. STUDY FUNDING/COMPETING INTEREST(S): No funding was received in support of this study. The authors have no conflicts of interest to disclose. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/complications , Female , Adolescent , Retrospective Studies , Hyperandrogenism/diagnosis , Practice Guidelines as Topic , Child , Ultrasonography , Insulin Resistance , Case-Control Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...