Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.052
Filter
1.
Syst Rev ; 13(1): 194, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049099

ABSTRACT

OBJECTIVES: Polycystic ovary syndrome (PCOS) is a prevalent and complex endocrine disorder that affects women of reproductive age. It has significant implications for female endocrine function, reproductive health, and metabolic disturbances, including insulin resistance, impaired glucose tolerance, and dyslipidemia. Studies have shown that decreased heart rate variability (HRV), a marker of autonomic dysfunction, is associated with adverse cardiovascular events. Recent research has focused on investigating autonomic function in PCOS, and some studies have suggested altered autonomic drive in these patients. The aim of this systematic review and meta-analysis was to evaluate cardiac autonomic function by analyzing HRV in women with PCOS. METHODS: This systematic review was prepared using PRISMA reporting guidelines. The databases searched were PubMed, Scopus, Web of Science, and Cochrane. Risk of Bias was assessed using ROBINS-I for non-RCTs. The GRADE approach was employed to evaluate the level of certainty in the evidence for each outcome. In order to identify the underlying cause of high heterogeneity, a subgroup analysis was conducted. Sensitivity analysis was checked. A random effect model was used and calculated a pooled standardized mean difference (SMD) with a 95% confidence interval (CI). RESULTS: Seventeen articles were included in the final analysis, varied in quality, ranging from a "low" to a "high risk of bias". Combined analyses indicated a notable decrease in HRV among individuals with PCOS compared to the control group. Significant changes were observed in SDNN (SMD: -0.763, 95%CI [-1.289 to -0.237], p=0.004), PNN50 (SMD: -1.245, 95%CI [-2.07, -0.419], p=0.003), LF/HF ratio (SMD: 0.670, 95%CI [0.248, 1.091], p=0.002), HFnu (SMD: -0.873, 95%CI [-1.257, -0.489], p=0.000), LFnu (SMD: 0.840, 95%CI [0.428, 1.251], p=0.000) and TP (SMD: -1.997, 95%CI [-3.306, -0.687], p=0.003). The heterogeneity was partially explained by types of study design. Subgroup analysis revealed significant alterations of HRV in normal-weighted and overweight PCOS cases. Conversely, no significant changes in HRV were observed in obese PCOS cases. CONCLUSION: The findings of this meta-analysis provide evidence suggesting diminished HRV in individuals with PCOS compared to non-PCOS control group.


Subject(s)
Cardiovascular Diseases , Heart Rate , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Humans , Female , Heart Rate/physiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Autonomic Nervous System/physiopathology
2.
Iran J Med Sci ; 49(6): 350-358, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38952638

ABSTRACT

Background: A combination of genetic and environmental factors contribute to the highly common, complex, and varied endocrine condition known as polycystic ovary syndrome (PCOS) in women. PCOS primarily affects women between the ages of 15 and 35 who are in the early to late stages of pregnancy. Thus, this study aimed to evaluate the serum levels of irisin, subfatin, and adropin in PCOS with and without obesity compared to the control group. Methods: The present cross-sectional study was conducted in 2022 at Al-Nahrain University/Department of Chemistry (Baghdad, Iraq). The serum levels of irisin, subfatin, and adropin were measured with the enzyme-linked immunosorbent assay (ELISA) method. Body mass index, lipid profile, insulin, fasting glucose, follicle-stimulating hormone, and luteinizing hormone levels were also evaluated. The data were analyzed using one-way analysis of variance (ANOVA) by GraphPad Prism software version 8.0.2. A P<0.05 was considered statistically significant. Results: The study population comprised PCOS patients (n=90, divided into 45 obese and 45 normal weight) and healthy women (n=30). According to the results, the serum levels of irisin were significantly higher (P<0.001) in obese and normal-weight PCOS patients than controls. While adropin and subfatin were significantly lower in PCOS than controls (P<0.001). Moreover, there are higher levels of serum insulin, fasting glucose, and luteinizing hormone in PCOS women than in healthy women. Conclusion: According to the findings, PCOS patients had a higher level of irisin than the controls. In addition, decreased subfatin and adropin levels were observed in PCOS patients compared with healthy women. Further research is required to confirm these results in the future.


Subject(s)
Fibronectins , Intercellular Signaling Peptides and Proteins , Obesity , Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/physiopathology , Adult , Fibronectins/blood , Fibronectins/analysis , Obesity/blood , Obesity/complications , Obesity/physiopathology , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/analysis , Cross-Sectional Studies , Young Adult , Blood Proteins/analysis , Peptides/blood , Peptides/analysis , Body Mass Index , Case-Control Studies , Adolescent
3.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062762

ABSTRACT

Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility.


Subject(s)
Gonadotropins , Ovarian Follicle , Polycystic Ovary Syndrome , Primary Ovarian Insufficiency , Animals , Female , Humans , Follicle Stimulating Hormone/metabolism , Gonadotropins/metabolism , Luteinizing Hormone/metabolism , Ovarian Follicle/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/etiology , Primary Ovarian Insufficiency/pathology
4.
Sci Rep ; 14(1): 13223, 2024 06 08.
Article in English | MEDLINE | ID: mdl-38851814

ABSTRACT

The aim of the study was to investigate the relation between thyroid autoimmunity (TAI), reflected as the presence of thyroid peroxidase antibodies (TPOAb), and parameters of ovarian reserve in women with type 1 diabetes (T1DM) and polycystic ovary syndrome (PCOS). We studied 83 euthyroid women with T1DM (age - 26 ± 5 years, BMI - 24 ± 3 kg/m2) - 12 with PCOS and positive TPOAb (PCOS + TPOAb), 29 with PCOS with negative TPOAb (PCOS + noTPOAb), 18 without PCOS with positive TPOAb (noPCOS + TPOAb), 24 without PCOS with negative TPOAb (noPCOS + noTPOAb). Serum concentrations of anti-Müllerian hormone (AMH), sex hormones, TSH, thyroid hormones and TPOAb were assessed. The prevalence of TAI was comparable between PCOS and noPCOS. We did not observe differences in hormonal profile or AMH concentration between two PCOS groups-PCOS + TPOAb and PCOS + noTPOAb (p > 0.05). Women with PCOS + TPOAb had lower FSH concentration and higher LH/FSH index than noPCOS + noTPOAb (p = 0.027; p = 0.019, respectively). Moreover, PCOS + TPOAb had lower oestradiol level than noPCOS + TPOAb (p = 0.041). AMH concentration was higher in both groups with PCOS, independent of TPOAb presence, than in noPCOS + noTPOAb (both p < 0.001). The presence of positive TPOAb titre was not related to the studied parameters of ovarian reserve - AMH and ovarian follicle number. In multiple linear regression analysis, the only significant predictor of AMH in the whole studied group with T1DM was total daily insulin dose U/kg (ß = - 0.264; p = 0.022). The presence of TAI did not affect the hormonal profile or ovarian reserve in women with T1DM with and without PCOS.


Subject(s)
Autoantibodies , Autoimmunity , Diabetes Mellitus, Type 1 , Ovarian Reserve , Polycystic Ovary Syndrome , Thyroid Gland , Humans , Female , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/physiopathology , Adult , Thyroid Gland/physiopathology , Thyroid Gland/immunology , Thyroid Gland/metabolism , Autoantibodies/blood , Autoantibodies/immunology , Young Adult , Anti-Mullerian Hormone/blood , Iodide Peroxidase/immunology
5.
Front Biosci (Landmark Ed) ; 29(6): 208, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38940030

ABSTRACT

Polycystic ovary syndrome (PCOS) is a prevalent reproductive, endocrine, and metabolic disease that affects 5-18% of women worldwide, with a rising incidence. Hyperandrogenemia and insulin resistance are two key pathophysiological factors that contribute to PCOS, both of which contribute to a variety of health issues such as menstrual irregularities, obesity, dysfunctional glucose and lipid homeostasis, infertility, mental disorders, and cardiovascular and cerebrovascular diseases. Despite ongoing studies, the origin and pathogenesis of PCOS remain elusive; there is also a clinical need for simpler, more effective, longer lasting, and more comprehensive treatments for women with PCOS. The gut-fat axis, a critical regulatory route for metabolism, endocrine function, and immune response, has received considerable interest in recent years in the research of the etiology and treatment of metabolic illnesses such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. The latest research in PCOS has revealed significant alterations in the homogeneity and phylogenetic diversity of the gut microbiota. Animal research using fecal microbiota transplantation has confirmed the importance of gut microbiota in regulating insulin sensitivity and sex hormone balance in PCOS. Furthermore, studies have shown a decrease in the volume and/or activity of brown adipose tissue (BAT) in PCOS patients, a change that alters adipokine release, leading to insulin resistance and hyperandrogenemia, aggravating PCOS progression. Given the function of BAT in increasing energy expenditure and alleviating metabolic parameters, efforts to activate BAT or induce browning of white adipose tissue have emerged as possible treatments for PCOS. Recent research has suggested that the gut microbiota can influence BAT creation and activity via metabolites such as short-chain fatty acids and bile acids, as well as the gut-brain axis. Cold exposure, healthy dieting, metformin, bariatric surgery, glucagon-like peptide 1 receptor agonists and melatonin have all been shown in basic and clinical studies to modulate BAT activity by influencing the gut microbiota, demonstrating significant clinical potential. However, more studies into the regulation mechanisms of the gut-BAT axis are required to produce more effective, comfortable, and safe tailored therapeutics for PCOS.


Subject(s)
Adipose Tissue, Brown , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/therapy , Polycystic Ovary Syndrome/physiopathology , Humans , Female , Gastrointestinal Microbiome/physiology , Adipose Tissue, Brown/metabolism , Animals , Insulin Resistance , Fecal Microbiota Transplantation , Obesity/microbiology , Obesity/metabolism , Obesity/therapy
6.
Hum Reprod ; 39(8): 1794-1803, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38867472

ABSTRACT

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


Subject(s)
Energy Metabolism , Obesity , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Female , Adult , Case-Control Studies , Obesity/metabolism , Obesity/complications , Obesity/physiopathology , Young Adult , Insulin Resistance , Body Mass Index , Basal Metabolism , Calorimetry, Indirect
7.
Gynecol Endocrinol ; 40(1): 2360077, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38818646

ABSTRACT

OBJECTIVE: To analyze differences in the menstrual pattern, age at menarche, and body mass index (BMI) in adolescents with Hypothalamic-Pituitary-Ovarian (HPO) axis immaturity and Polycystic Ovary Syndrome (PCOS) through a systematic review and meta-analysis. METHODS: The PubMed, EMBASE, Web of Science, Virtual Health Library, Scopus databases were searched using combinations of descriptors. Study quality was assessed using the Newcastle-Ottawa Scale. For data analysis, the results were grouped into PCOS group and NPCOS group (HPO axis immaturity). We performed a meta-analysis of raw data and the inverse variance method, employing the standardized mean difference, of the age at menarche and BMI of adolescents. RESULTS: Participants totaled 1,718 from nine selected studies. The meta-analysis showed that the PCOS group had a higher BMI than the NPCOS group (SMD 0.334; CI95% 0.073 - 0.595; p = .012). The degree of heterogeneity of the studies was approximately 40%. No significant difference in age at menarche (SMD - 0.027; CI95% -0.227 - 0.172; p = 0.790) and menstrual patterns was found, but amenorrhea was described only in adolescents with PCOS. CONCLUSIONS: The main characteristic in menstrual pattern that differentiated PCOS patients from girls with HPO axis immaturity was amenorrhea. Also, the BMI of PCOS patients was nearly one third higher than that of adolescents with HPO axis immaturity.


Subject(s)
Body Mass Index , Hypothalamo-Hypophyseal System , Menarche , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Female , Adolescent , Menarche/physiology , Hypothalamo-Hypophyseal System/physiopathology , Ovary/physiopathology , Menstrual Cycle/physiology
8.
Gynecol Endocrinol ; 40(1): 2352139, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38733361

ABSTRACT

OBJECTIVE: The main purpose of this systematic review and meta-analysis was to investigate the diagnostic value of ultrasound elastography in the evaluation of polycystic ovary syndrome (PCOS). METHODS: A comprehensive and methodical investigation was carried out in the databases of PubMed, EMBASE, Cochrane, Scopus, Web of Science, and China National Knowledge Infrastructure, covering the entire duration of these databases until October 18, 2023. The primary purpose of this research was to evaluate and contrast ovarian tissue elasticity in people with and without PCOS. The elasticity of ovarian tissue was quantified using standardized mean difference (SMD). RESULTS: A total of eight studies were ultimately selected for systematic evaluation and meta-analysis. Five studies used shear wave elastography (SWE) as a diagnostic tool, and it was discovered that women with PCOS had higher levels of ovarian shear wave elasticity than their healthy counterparts. The SMD was determined to be 1.86 kilopascal (95% CI: 1.27 to 2.44). Three studies were conducted using strain elastography (SE) to compare the ovarian strain ratio of patients with PCOS to that of a healthy control group. The SMD for the PCOS group was 2.07 (95% CI: 1.79 to 2.34), which indicated that the ovarian strain ratio was significantly higher in that group. CONCLUSION: This systematic review and meta-analysis found that women with PCOS had stiffer ovarian tissue than women without the disorder. Ultrasound elastography may provide clinicians with value beyond 2D ultrasound in the diagnosis of PCOS.


Subject(s)
Elasticity Imaging Techniques , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/diagnostic imaging , Polycystic Ovary Syndrome/physiopathology , Humans , Elasticity Imaging Techniques/methods , Female , Ovary/diagnostic imaging , Elasticity
9.
Zhongguo Zhong Yao Za Zhi ; 49(8): 2006-2015, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38812217

ABSTRACT

This study aims to observe the efficacy and safety of Bushen Culuan Formula in the treatment of infertility caused by polycystic ovary syndrome(PCOS) and to explore the mechanism using metabolomics. Ninety-four patients with infertility caused by PCOS with the syndrome of kidney deficiency and blood stasis were selected and assigned into treatment and control groups(n=47). The basal body temperature(BBT) was measured, and B-ultrasonography was employed to monitor follicles, ovarian volume, endometrium, ovulation, and pregnancy. The serum levels of sex hormones including follicle-stimulating hormone(FSH), luteinizing hormone(LH), prolactin(PRL), estradiol(E_2), progestin(P), testosterone(T), free testosterone(FT), androstenedione(A2), inhibin B(INHB), and anti-Müllerian hormone(AMH) were measured. The coagulation function, traditional Chinese medicine(TCM) symptom scores, blood and urine routine, liver and kidney functions and other safety indicators were determined. Metabolomics was employed to comparatively analyze the serum metabolites of 26 patients(13 patients in each group) in the clinical study. The results showed that the total response rate and pregnancy rate of the treatment group were higher than those of the control group(P<0.001), suggesting that Bushen Culuan Formula regulated the sex hormones and ovarian function. Specifically, it reduced the levels of LH, T, FT, A2, and INHB(P<0.05 or P<0.01) and the LH/FSH ratio(P<0.05), elevated the level of P(P<0.05), promoted ovulation, increased endothelial thickness, and lowered TCM symptom scores without causing adverse reactions. A total of 24 differential metabolites were screened by metabolomics, and there were correlations between sex hormones and differential metabolites in the PCOS-induced infertility patients with kidney deficiency and blood stasis. In conclusion, Bushen Culuan Formula may regulate hormone levels through lipid and amino acid metabolism.


Subject(s)
Drugs, Chinese Herbal , Infertility, Female , Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Drugs, Chinese Herbal/administration & dosage , Adult , Infertility, Female/drug therapy , Infertility, Female/etiology , Infertility, Female/physiopathology , Young Adult , Pregnancy , Luteinizing Hormone/blood
10.
Endocrinol Diabetes Metab ; 7(3): e490, 2024 May.
Article in English | MEDLINE | ID: mdl-38769719

ABSTRACT

OBJECTIVE: This study aimed to investigate whether polycystic ovary syndrome (PCOS) status changes the association between insulin resistance (IR) indices and liver function parameters among women. METHODS: This is a cross-sectional, population-based study. We selected 1101 subjects aged ≥20 years from participants of Tehran Lipid and Glucose Study (TLGS). All of them had known the status of PCOS, and all variables were related to the IR indices and liver function parameters. The main outcome measures were TG/HDL-C and triglyceride-glucose (TyG) and liver function parameters (hepatic steatosis index [HSI], alanine transaminase [ALT] and aspartate transaminase [AST]). RESULT: In the present study, there was no significant difference between the PCOS and the non-PCOS regarding the presence of liver function abnormalities. A model adjusted by age and BMI showed that the upper tertile of TyG index was positively associated with high AST (OR = 3.04 [95% CI: 1.20-7.68], p < 0.05), high ALT (4.76 [3.07-7.36], p < 0.05) and high HSI (8.44 [1.82-39.17], p < 0.05). Although the history of diabetes had a positive impact on elevated AST (1.66 [1.15, 2.40], p < 0.05), the third tertile of TG/HDL-C was associated with increased odds of elevated ALT (3.35 [2.21-5.06]) and HSI (6.55 [1.17-36.46]), whereas the second tertile of TG/HDL-C (OR = 2.65, CI 95%: 1.74-4.03) was also positively associated with elevated ALT. PCOS had no significant association with elevated liver function tests. CONCLUSION: The highest tertile of TyG index and the TG/HDL-C ratio as a surrogate of IR might play a role in detecting abnormalities of liver function parameters among women. However, PCOS status cannot change the association between IR and liver dysfunction.


Subject(s)
Alanine Transaminase , Insulin Resistance , Liver Function Tests , Liver , Polycystic Ovary Syndrome , Triglycerides , Humans , Female , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Adult , Cross-Sectional Studies , Liver/metabolism , Triglycerides/blood , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Iran/epidemiology , Blood Glucose/metabolism , Young Adult , Cholesterol, HDL/blood , Fatty Liver/etiology , Fatty Liver/physiopathology
11.
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
12.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732117

ABSTRACT

Glomerular hyperfiltration (GH) has been reported to be higher in women with polycystic ovary syndrome (PCOS) and is an independent risk factor for renal function deterioration, metabolic, and cardiovascular disease. The aim of this study was to determine GH in type A PCOS subjects and to identify whether inflammatory markers, markers of CKD, renal tubule injury markers, and complement system proteins were associated. In addition, a secondary cohort study was performed to determine if the eGFR had altered over time. In this comparative cross-sectional analysis, demographic, metabolic, and proteomic data from Caucasian women aged 18-40 years from a PCOS Biobank (137 with PCOS, 97 controls) was analyzed. Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement was undertaken for inflammatory proteins, serum markers of chronic kidney disease (CKD), tubular renal injury markers, and complement system proteins. A total of 44.5% of the PCOS cohort had GH (eGFR ≥ 126 mL/min/1.73 m2 (n = 55)), and 12% (n = 17) eGFR ≥ 142 mL/min/1.73 m2 (super-GH(SGH)). PCOS-GH women were younger and had lower creatinine and urea versus PCOS-nonGH. C-reactive protein (CRP), white cell count (WCC), and systolic blood pressure (SBP) were higher in PCOS versus controls, but CRP correlated only with PCOS-SGH alone. Complement protein changes were seen between controls and PCOS-nonGH, and decay-accelerator factor (DAF) was decreased between PCOS-nonGH and PCOS-GSGH (p < 0.05). CRP correlated with eGFR in the PCOS-SGH group, but not with other inflammatory or complement parameters. Cystatin-c (a marker of CKD) was reduced between PCOS-nonGH and PCOS-GSGH (p < 0.05). No differences in tubular renal injury markers were found. A secondary cohort notes review of the biobank subjects 8.2-9.6 years later showed a reduction in eGFR: controls -6.4 ± 12.6 mL/min/1.73 m2 (-5.3 ± 11.5%; decrease 0.65%/year); PCOS-nonGH -11.3 ± 13.7 mL/min/1.73 m2 (-9.7 ± 12.2%; p < 0.05, decrease 1%/year); PCOS-GH (eGFR 126-140 mL/min/17.3 m2) -27.1 ± 12.8 mL/min/1.73 m2 (-19.1 ± 8.7%; p < 0.0001, decrease 2%/year); PCOS-SGH (eGFR ≥ 142 mL/min/17.3 m2) -33.7 ± 8.9 mL/min/17.3 m2 (-22.8 ± 6.0%; p < 0.0001, decrease 3.5%/year); PCOS-nonGH eGFR versus PCOS-GH and PCOS-SGH, p < 0.001; no difference PCOS-GH versus PCOS-SGH. GH was associated with PCOS and did not appear mediated through tubular renal injury; however, cystatin-c and DAF were decreased, and CRP correlated positively with PCOS-SGH, suggesting inflammation may be involved at higher GH. There were progressive eGFR decrements for PCOS-nonGH, PCOS-GH, and PCOS-SGH in the follow-up period which, in the presence of additional factors affecting renal function, may be clinically important in the development of CKD in PCOS.


Subject(s)
Biomarkers , Glomerular Filtration Rate , Polycystic Ovary Syndrome , Renal Insufficiency, Chronic , Humans , Female , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/blood , Adult , Cross-Sectional Studies , Biomarkers/blood , Young Adult , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/etiology , Adolescent , C-Reactive Protein/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/metabolism
13.
Mol Biol Rep ; 51(1): 631, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722405

ABSTRACT

Adipokines are now well-known to regulate reproduction. Visfatin is an adipokine expressed in the hypothalamus, pituitary, ovary, uterus, and placenta of different species, and since it has been found to modulate the endocrine secretion of the hypothalamus, pituitary gland and ovary, it may be considered a novel regulator of female reproduction. Although the majority of the literature explored its role in ovarian regulation, visfatin has also been shown to regulate uterine remodeling, endometrial receptivity and embryo development, and its expression in the uterus is steroid dependent. Like other adipokines, visfatin expression and levels are deregulated in pathological conditions including polycystic ovary syndrome. Thus, the present mini-review focuses on the role of visfatin in female reproduction under both physiological and pathological conditions.


Subject(s)
Nicotinamide Phosphoribosyltransferase , Polycystic Ovary Syndrome , Reproduction , Female , Humans , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Reproduction/physiology , Reproduction/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Animals , Ovary/metabolism , Uterus/metabolism , Cytokines/metabolism , Pregnancy , Adipokines/metabolism
14.
Maturitas ; 185: 107982, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38579578

ABSTRACT

OBJECTIVE: The potential effects of polycystic ovary syndrome (PCOS) on the musculoskeletal system are not well established. We examined the musculoskeletal system in women with PCOS in their late reproductive years. STUDY-DESIGN: This cross-sectional study included 34 women with PCOS and 32 control women matched for age and body mass index (BMI). MAIN OUTCOME MEASURES: Dual-energy x-ray absorptiometry (DXA) was used for body composition analysis and cross-sectional areas and fat fraction of muscles were assessed by magnetic resonance imaging-proton density fat fraction (MRI-PDFF) of the abdomen and thigh. Muscle strength was measured using an isokinetic dynamometer. RESULTS: The mean age of the PCOS group was 43 ± 3.7 years and of the control group 42.2 ± 3.5 years. Testosterone, free androgen index, and fasting insulin were higher in PCOS patients than controls (p < 0.001, p = 0.001 and p = 0.032, respectively). Patients and controls had similar values for total abdominal muscle area (TAMA), paraspinal muscle area, thigh muscle area, vertebral MRI-PDFF, thigh and paraspinal muscle MRI-PDFF. There was no difference in DXA-derived muscle and bone composition between the two groups. Body composition parameters measured by MRI and DXA were correlated with BMI and fasting insulin levels, but not with androgen levels in both groups. Subgroup analyses showed that PCOS women with obesity had higher TAMA than controls with obesity (p = 0.012). Apart than higher 60°/sec knee extensor average power in nonobese PCOS (p = 0.049), no difference in muscle mechanical function was detected between PCOS patients and controls. CONCLUSION: Musculoskeletal composition and function are similar in PCOS patients and healthy women in late reproductive years. Body composition is linked with obesity and insulin resistance rather than hyperandrogenemia.


Subject(s)
Absorptiometry, Photon , Body Composition , Magnetic Resonance Imaging , Muscle Strength , Muscle, Skeletal , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/physiopathology , Female , Adult , Cross-Sectional Studies , Muscle, Skeletal/physiopathology , Muscle, Skeletal/diagnostic imaging , Body Mass Index , Testosterone/blood , Case-Control Studies , Middle Aged , Aging/physiology , Bone Density , Insulin/blood , Thigh , Abdominal Muscles/physiopathology , Abdominal Muscles/diagnostic imaging , Bone and Bones/physiopathology , Bone and Bones/diagnostic imaging
15.
Semin Reprod Med ; 42(1): 34-40, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38608673

ABSTRACT

In recent years, the expanding roles of anti-Müllerian hormone (AMH) in various aspects of reproductive health have attracted significant attention. Initially recognized for its classical role in male sexual differentiation, AMH is produced postnatally by the Sertoli cells in the male testes and by the granulosa cells in the female ovaries. Traditionally, it was believed to primarily influence gonadal development and function. However, research over the last decade has unveiled novel actions of AMH beyond the gonads, specifically all along the hypothalamic-pituitary-gonadal axis. This review will focus on the emerging roles of AMH within the hypothalamus and discusses its potential implications in reproductive physiology. Additionally, recent preclinical and clinical studies have suggested that elevated levels of AMH may disrupt the hypothalamic network regulating reproduction, which could contribute to the central pathophysiology of polycystic ovary syndrome. These findings underscore the intricate interplay between AMH and the neuroendocrine system, offering new avenues for understanding the mechanisms underlying fertility and reproductive disorders.


Subject(s)
Anti-Mullerian Hormone , Fertility , Hypothalamus , Anti-Mullerian Hormone/metabolism , Humans , Fertility/physiology , Female , Animals , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Ovary/metabolism , Ovary/physiology , Signal Transduction
16.
Curr Cardiol Rep ; 26(6): 483-495, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38568339

ABSTRACT

PURPOSE OF REVIEW: Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder in women of reproductive age. It has been associated with metabolic, reproductive, and psychiatric disorders. Despite its association with insulin resistance (IR) and cardiovascular disease (CVD) risk factors, the association between PCOS and CVD outcomes has been conflicting. This review reports the updated evidence between PCOS, insulin resistance, and CVD events. RECENT FINDINGS: IR is highly prevalent occurring in 50 to 95% of general and obese PCOS women. The etiology of PCOS involves IR and hyperandrogenism, which lead to CVD risk factors, subclinical CVD, and CVD outcomes. Multiple studies including meta-analysis confirmed a strong association between PCOS and CVD events including ischemic heart disease, stroke, atrial fibrillation, and diabetes, particularly among premenopausal women, and these associations were mediated by metabolic abnormalities. PCOS is highly familial and has substantial CVD risk and transgenerational effects regardless of obesity. A personalized approach to the CVD risk assessment and management of symptom manifestations should be conducted according to its phenotypes. Lifestyle modifications and reduction in environmental stressors should be encouraged for CVD prevention among PCOS women.


Subject(s)
Cardiovascular Diseases , Insulin Resistance , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/physiopathology , Female , Cardiovascular Diseases/etiology , Obesity/complications , Obesity/physiopathology , Risk Factors , Risk Assessment , Heart Disease Risk Factors , Prevalence , Hyperandrogenism/complications , Hyperandrogenism/physiopathology
18.
J Endocrinol Invest ; 47(7): 1-22, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38485896

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is the most common feminine endocrine disorder, characterized by androgen excess, ovulatory dysfunction, and polycystic ovarian morphology. The negative impact of symptoms on the quality of life (QoL) of patients is still not clear. PURPOSE: The present review aimed at studying the impact of the symptoms, the psychological symptoms, and brain alterations in women with PCOS. METHODS: A systematic search was undertaken for studies that assessed the impact of PCOS symptoms on QoL, psychological symptoms, and brain alterations in PCOS patients. RESULTS: Most of the information about QoL came from psychometric studies, which used culture-based questionnaires. Alterations of sleep quality, body image, and mood disorders can negatively affect the QoL of the patients. Sexual satisfaction and desire were affected by PCOS. Brain imaging studies showed functional alterations that are associated with impairments of visuospatial working memory, episodic and verbal memory, attention, and executive function. CONCLUSIONS: Several factors can negatively influence the quality of life of the patients, and they are directly related to hyperandrogenism and the risk of infertility. In particular, obesity, hirsutism, acne, and the fear of infertility can have a direct impact on self-esteem and sexual function. Metabolic and psychiatric comorbidities, such as mood, anxiety, and eating disorders, can affect the well-being of the patients. Moreover, specific cognitive alterations, such as impairments in attention and memory, can limit PCOS patients in a series of aspects of daily life.


Subject(s)
Brain , Polycystic Ovary Syndrome , Quality of Life , Humans , Polycystic Ovary Syndrome/psychology , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/complications , Female , Brain/diagnostic imaging , Brain/physiopathology
19.
J Obstet Gynaecol Res ; 50(5): 800-808, 2024 May.
Article in English | MEDLINE | ID: mdl-38412992

ABSTRACT

The follicular microenvironment is crucial for normal ovarian function, and intra-ovarian factors, in coordination with gonadotropins, contribute to its regulation. Recent research has revealed that the accumulation of senescent cells worsens the adverse environment of various tissues and plays critical roles in chronological aging and various pathological conditions. Cellular senescence involves cell-cycle arrest, a senescence-associated secretory phenotype (SASP), macromolecular damage, and dysmetabolism. In this review, I summarize the latest knowledge regarding the role of cellular senescence in pathological conditions in the ovary, in the context of reproduction. Specifically, cellular senescence is known to impair follicular and oocyte health in cisplatin- and cyclophosphamide-induced primary ovarian insufficiency and to contribute to the pathogenesis of polycystic ovary syndrome (PCOS). In addition, cellular senescence is induced during the decline in ovarian reserve that is associated with chronological aging, endometriosis, psychological stress, and obesity, but it remains unclear whether it plays a causative role in these conditions. Finally, I discuss the potential for use of cellular senescence as a novel therapeutic target. The modification of SASP using a senomorphic and/or the elimination of senescent cells using a senolytic represent promising therapeutic strategies. Further elucidation of the role of cellular senescence in the effects of various insults on ovarian reserve, including chronological aging, as well as in pathogenesis of ovarian pathologies, including PCOS, may facilitate a new era of reproductive medicine.


Subject(s)
Cellular Senescence , Humans , Female , Cellular Senescence/physiology , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/metabolism , Primary Ovarian Insufficiency/physiopathology , Ovary/physiopathology , Ovary/physiology , Ovarian Diseases/physiopathology , Aging/physiology , Ovarian Reserve/physiology
20.
Fertil Steril ; 121(6): 1010-1019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38307452

ABSTRACT

OBJECTIVE: To derive and internally validate a clinical prediction model for live birth (LB) in women with polycystic ovary syndrome (PCOS) undergoing in vitro fertilization (IVF). DESIGN: Retrospective cohort study. SETTING: Four academic reproductive endocrinology clinics. PATIENTS: A total of 207 women with PCOS confirmed using Rotterdam criteria undergoing their first fresh IVF cycle. INTERVENTIONS: Not applicable. MAIN OUTCOME MEASURE: The primary outcome was cumulative LB per IVF cycle start. This included any LB that resulted from either fresh embryo transfer or any subsequent frozen embryo transfer from embryos obtained at the index oocyte retrieval. A prediction model was derived using multivariable logistic regression. Covariates considered for inclusion in the prediction model included demographic characteristics, medical history, and prior fertility treatment. Predicted probabilities for LB were calculated using the prediction model which included the 90% shrinkage factor for each adjusted odds ratio. RESULTS: The final model, on the basis of maximization of the area under the receiver operating characteristic curve, included age < 35 years, White race, presence of polycystic ovaries on ultrasound (polycystic ovary morphology), normal body mass index (<25 kg/m2), being metabolically healthy (no metabolic risk factors), and being a nonresponder to ovulation induction agents including letrozole and clomiphene citrate. The area under the receiver operating characteristic curve score for the model was 0.68 (95% confidence interval [CI]: 0.60, 0.77). Predicted probabilities of LB ranged from 8.1% (95% CI: 2.8, 21.5) for a woman who had no favorable predictors to 74.2% (95% CI: 59.5, 84.9) for a woman who had all favorable predictors. CONCLUSION: Our study demonstrated that, in addition to anovulation, the underlying pathophysiology and associated comorbidities alter the likelihood of a successful pregnancy in women with PCOS undergoing IVF. Further validation of this model is needed before it can serve as a tool to personalize prediction estimates for the probability of LB in women with PCOS.


Subject(s)
Fertilization in Vitro , Infertility, Female , Live Birth , Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/therapy , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/complications , Fertilization in Vitro/methods , Adult , Pregnancy , Retrospective Studies , Infertility, Female/therapy , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Infertility, Female/epidemiology , Treatment Outcome , Embryo Transfer/methods , Risk Factors , Pregnancy Rate , Risk Assessment , Reproducibility of Results , Ovulation Induction/methods , Predictive Value of Tests , Decision Support Techniques
SELECTION OF CITATIONS
SEARCH DETAIL