1
|
Zhang X, Liu J, Bai C, Fan Y, Song H, Huang Z, Li Y, Luo T. Palmitic acid enhances the sensitivity of ferroptosis via endoplasmic reticulum stress mediated the ATF4/TXNIP Axis in polycystic ovary syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156777. [PMID: 40393214 DOI: 10.1016/j.phymed.2025.156777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Palmitic acid (PA), the most prevalent saturated fatty acid in humans, is closely associated with ovarian dysfunction. Elevated PA levels in the follicular fluid of patients with polycystic ovary syndrome (PCOS) are correlated with the outcomes of assisted reproductive technology (ART), though the underlying mechanism remains unclear. METHODS Multi-omics analysis identified PA and TXNIP as potential pathogenic factors in PCOS. CCK-8 and apoptosis assay were conducted to detect the cytotoxicity of PA. To further investigate the molecular mechanisms underlying PA-induced ferroptosis, we established COV434 cell models with both TXNIP overexpression and knockdown. Transmission electron microscopy (TEM), western blot (WB), ELISA assays, and flow cytometry were employed to assess ferroptosis-related markers. A PCOS mouse model was also developed, and histopathological staining, TEM, ELISA, and WB were performed to evaluate clinical parameters related to PCOS ovarian ferroptosis levels. To clarify the targeting relationship between ATF4 and TXNIP, we utilized luciferase reporter gene assays, chromatin immunoprecipitation (ChIP), and RT-qPCR for a comprehensive analysis. RESULTS In vivo and in vitro, PA enhanced the sensitivity of PCOS ovarian ferroptosis. The protein levels of TXNIP and ACSL4 were upregulated in both PCOS patients and mouse models after PA treatment. PA also induces the expression of the ferroptosis inhibitor SLC7A11 as part of an adaptive response. Elevated intracellular ROS levels, increased MDA content, decreased GSH/GSSG ratios, elevated ferrous iron levels, and TEM findings collectively indicated that PA induces ferroptosis in KGN/COV434 cells. The ER stress inhibitor 4-PBA reduces PA-induced ferroptosis in PCOS ovaries by suppressing ER stress, thereby improving PA-induced PCOS-like traits. Moreover, the UPR gene ATF4 regulates cellular ferroptosis by activating the transcriptional expression of TXNIP. CONCLUSION PA stimulated ovarian ferroptosis by activating ER stress, a process mediated by the ATF4/TXNIP axis, which might represent a potential mechanism underlying the progression of PCOS. The application of ER stress inhibitors improved PCOS traits by reducing the sensitivity of ovarian ferroptosis.
Collapse
Affiliation(s)
- Xumin Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Jianrong Liu
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China; Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China.
| | - Chunmei Bai
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Yanxin Fan
- Department of Orthopedics, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtasi Street, Taiyuan 030012, Shanxi, PR China
| | - Haixia Song
- Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Ziwei Huang
- Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Yang Li
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Ting Luo
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| |
Collapse
|
2
|
Ding N, Chen YR, Jia RJ, Lu XZ, Xie SL, Shang HL, Shuai JG. Interaction between interleukin 10 ( IL-10) gene polymorphisms and obesity on susceptibility to polycystic ovary syndrome in Chinese women. Endocr Res 2025:1-7. [PMID: 40562065 DOI: 10.1080/07435800.2025.2521386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/20/2025] [Accepted: 06/12/2025] [Indexed: 06/28/2025]
Abstract
OBJECTIVES The pathogenesis of polycystic ovary syndrome (PCOS) was complex, and the incident PCOS involves both genetic and environmental factors. However, no study focused on the synergistic effect between interleukin 10 (IL-10) gene and obesity on PCOS risk yet. This study aimed to evaluate the correlation between IL-10 gene single nucleotide polymorphisms (SNPs) and PCOS susceptibility and impact of the interaction between IL-10 gene and obesity on PCOS risk. METHODS A total of 540 participants consisted of 180 PCOS patients and 360 normal controls were enrolled in this study. Logistic regression model was employed to evaluate the association between IL-10 gene polymorphisms and PCOS susceptibility, odds ratios (ORs) and 95% confidence interval (CI) were calculated. Generalized multifactor dimensionality reduction (GMDR) was employed to screen the IL-10 gene-obesity interaction. RESULTS Logistic regression also indicated that rs1800896-G allele was statistically significant correlated with increased risk of PCOS, the ORs (rs (95%CI) for AG, GG and AG+GG genotype was 1.75 (1.21-2.33), 1.93 (1.17-2.72) and 1.79 (1.26-2.35), respectively. However, no significant difference was observed on the distribution of genotypes and alleles within rs1800890, rs1800871, rs1800872 between PCOS patients and normal controls (all p values > 0.05). GMDR model found a significant interaction combination (two-locus model with p = 0.001) between rs1800896 and obesity, the cross-validation consistency was 10/10 and the prediction error was 0.641. Compared with those non-obese participants with rs1800896-AA genotype, OR (95% CI) was 1.62 (1.14-2.12), 1.46 (1.02-1.95) for non-obese participants with rs1800896-AG or GG genotype, obese participants with rs1800896-AA genotype, and obese participants with rs1800896-AG or GG genotype have the highest PCOS risk, OR (95% CI) = 3.58 (1.81-5.41), after covariates adjusting. CONCLUSIONS We found that rs1800896-G allele, gene-environment interaction between rs1800896 and obesity were all correlated with increased PCOS risk.
Collapse
Affiliation(s)
- Ning Ding
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| | - Yi-Rou Chen
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| | - Rui-Juan Jia
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| | - Xi-Zhou Lu
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| | - Shu-Lin Xie
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| | - Hui-Ling Shang
- Department of Gynecology, Foshan Women and Child Hospital, Foshan city, Guangdong province, China
| | - Jian-Gang Shuai
- Department of Gynecology, The Second People's Hospital of Foshan, Foshan city, Guangdong province, China
| |
Collapse
|
3
|
Bauer R, Parker C, Gorsic LK, Hayes MG, Kunselman AR, Legro RS, Welt CK, Urbanek M. Rare Variation in LMNA Underlies Polycystic Ovary Syndrome Pathogenesis in 2 Independent Cohorts. J Clin Endocrinol Metab 2025; 110:e2217-e2232. [PMID: 39484826 PMCID: PMC12187200 DOI: 10.1210/clinem/dgae761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is a common, heritable endocrinopathy that is a common cause of anovulatory infertility in reproductive age women. Variants in LMNA cause partial lipodystrophy, a syndrome with overlapping features to PCOS. OBJECTIVE We tested the hypothesis that rare variation in LMNA contributes to PCOS pathogenesis and selects a lipodystrophy-like subtype of PCOS. METHODS We sequenced LMNA by targeted sequencing a Discovery cohort of 811 PCOS patients and 164 healthy controls. We then analyzed LMNA from whole-exome sequencing of a Replication cohort of 718 PCOS patients and 281 healthy controls. We evaluated variation in the LMNA gene and hormone and lipid profiles of participants. RESULTS In the Discovery cohort, we identified 8 missense variants in 15/811 cases, and 1 variant in 1/172 reproductively healthy controls. There is strong evidence for association between the variants and PCOS compared to gnomAD non-Finnish European population controls (χ2 = 17, P = 3.7 × 10-5, OR = 2.9). In the Replication cohort, we identified 11 unique variants in 15/718 cases, and 1 variant in 281 reproductively healthy controls. Again, there is strong evidence for association with population controls (χ2 = 30.5, P = 3.4 × 10-8, OR = 4.0). In both the Discovery and Replication cohorts, variants in LMNA identify women with PCOS with high triglycerides and extreme insulin resistance. CONCLUSION Rare missense variation in LMNA is reproducibly associated with PCOS and identifies some individuals with lipodystrophy-like features. The overlap between this PCOS phenotype and genetic partial lipodystrophy syndromes warrants further investigation into additional lipodystrophy genes and their potential in PCOS etiology.
Collapse
Affiliation(s)
- Rosemary Bauer
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Reproductive Science, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Parker
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lidija K Gorsic
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Anthropology, Northwestern University, Evanston, IL 60208, USA
| | - Allen R Kunselman
- Public Health Sciences, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Richard S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism, and Diabetes, University of Utah, Salt Lake City, UT 84132, USA
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Reproductive Science, Northwestern University, Chicago, IL 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
4
|
Aksun S, Karaçoban L, Idilman I, Yildiz BO. Impact of oral contraceptive use on muscle mass and strength in women with PCOS. Endocrine 2025:10.1007/s12020-025-04305-9. [PMID: 40514618 DOI: 10.1007/s12020-025-04305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is characterized by androgen excess and ovulatory dysfunction and appears to be associated with alterations in muscle mass and function. The study aims to investigate whether oral contraceptive (OC) use affects muscle mass and strength in women with PCOS. METHODS Twenty women with PCOS (median age 20.5 years and BMI 26.1 kg/m2) and 20 age- and BMI-matched healthy controls were included. Clinical, hormonal, and biochemical assessments were conducted along with body composition analyses using magnetic resonance imaging (MRI) proton density fat fraction (PDFF%) and muscular strength assessment by isokinetic dynamometry. In women with PCOS, measurements were repeated after at least three cycles of OC therapy. RESULTS At baseline, women with PCOS exhibited significantly higher levels of total testosterone, free androgen index (FAI), and homeostasis model assessment of insulin resistance (HOMA-IR) levels compared to healthy controls (p < 0.001, p = 0.001, p = 0.004, respectively). PCOS group also showed significantly higher average power (AvP) of knee extensors at 60°/sec (p = 0.002). AvP correlated positively with total testosterone and FAI levels in the whole study group (r = 0.450, p = 0.004, r = 0.318, p = 0.045, respectively). Following OC therapy, testosterone levels and FAI decreased (p = 0.02 and p < 0.001, respectively); whereas thigh muscle mass or lower limb strength remained unchanged. CONCLUSIONS Short-term OC use in women with PCOS led to a reduction in androgen excess without measurable effects on muscle composition or strength. These findings suggest that muscle function and composition remain stable over the short term, despite hormonal modulation. Further research is required to understand how long-term management strategies for PCOS might affect muscle mass and function.
Collapse
Affiliation(s)
- Seren Aksun
- Hacettepe University School of Medicine, Department of Internal Medicine, Ankara, Turkey
- Hacettepe University School of Medicine, Department of Internal Medicine, Division of Endocrinology and Metabolism, Ankara, Turkey
| | - Levend Karaçoban
- Hacettepe University School of Medicine, Department of Sports Medicine, Ankara, Turkey
| | - Ilkay Idilman
- Hacettepe University School of Medicine, Department of Radiology, Ankara, Turkey
| | - Bulent O Yildiz
- Hacettepe University School of Medicine, Department of Internal Medicine, Division of Endocrinology and Metabolism, Ankara, Turkey.
| |
Collapse
|
5
|
Liu Q, Hu KL, Shi J, Yang S, Chu Y, Wang H, Zhang Y, Yue J, Hao G, Wu X, Zhao J, Li R. Association between anti-Müllerian hormone levels during pregnancy and pregnancy outcomes in infertile patients undergoing in vitro fertilisation/intracytoplasmic sperm injection: protocol for a multicentre prospective cohort study. BMJ Open 2025; 15:e093543. [PMID: 40499959 PMCID: PMC12161335 DOI: 10.1136/bmjopen-2024-093543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 05/20/2025] [Indexed: 06/18/2025] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS), recognised as the predominant aetiological factor in ovulatory dysfunction-related infertility, accounts for approximately 70% of anovulatory infertility cases. Patients with PCOS have significantly higher anti-Müllerian hormone (AMH) levels than their counterparts undergoing in vitro fertilisation (IVF) for non-PCOS indications (eg, male/tubal factors). Several studies have suggested that a high AMH level is associated with adverse pregnancy outcomes in IVF, particularly preterm delivery. However, most of these studies are retrospective studies, and their results are inconsistent. The majority of AMH measurements are conducted before pregnancy; however, AMH levels fluctuate dynamically during pregnancy. There is a pressing need for a well-structured prospective study to definitively establish whether high AMH levels during pregnancy are associated with IVF/intracytoplasmic sperm injection (ICSI) pregnancy outcomes in PCOS patients. METHODS AND ANALYSIS This prospective cohort study will be conducted at four reproductive medicine centres. The plan is to enrol 1,320 PCOS patients and 1320 non-PCOS women who undergo IVF/ICSI and achieve singleton clinical pregnancies. Serum samples will be collected at about 6 weeks of gestation to measure the serum AMH level. Follow-up visits will be conducted at 12, 28 and 37 weeks of gestation, delivery and 6 weeks after delivery to obtain information about pregnancy outcomes and complications. The primary outcome is preterm delivery. ETHICS AND DISSEMINATION The study was approved by the Medical Research Ethics Committee of Peking University Third Hospital (M2022618). Informed consent will be obtained from all patients. The results of this clinical study will be presented at scientific conferences and submitted to a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2300068554.
Collapse
Affiliation(s)
- Qiong Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Kai-Lun Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Jing Shi
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Shulin Yang
- Center for Reproductive Medicine, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Wuhan, Hubei, China
| | - Yifan Chu
- Center for Reproductive Medicine, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Wuhan, Hubei, China
| | - Honghong Wang
- Center for Reproductive Medicine, Children's Hospital of Shanxi, Taiyuan, Shanxi, China
| | - Yiwen Zhang
- Center for Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Yue
- Center for Reproductive Medicine, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Wuhan, Hubei, China
| | - Guimin Hao
- Center for Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xueqing Wu
- Center for Reproductive Medicine, Children's Hospital of Shanxi, Taiyuan, Shanxi, China
| | - Jie Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
6
|
Olaniyi KS, Okara DO, Areloegbe SE. Suppression of adipose TGF-β1 by probiotics alleviates metabolic disturbance in experimentally induced PCOS. Prostaglandins Other Lipid Mediat 2025; 178:106989. [PMID: 40154941 DOI: 10.1016/j.prostaglandins.2025.106989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/08/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is critically characterized with metabolic and endocrine dysfunctions, precipitating metabolic syndrome and infertility in reproductive aged women. Adipose tissue dysfunction has been implicated in the pathogenesis of metabolic syndrome, including in PCOS individuals. Probiotics are healthy bacteria in the gut that regulate metabolic health. However, the impact of probiotics on adipose-driven metabolic syndrome has not been reported. The present study therefore hypothesized that probiotics would attenuates metabolic disturbance in experimental PCOS rat model, probably by suppression of TGF-β1. MATERIALS AND METHODS Eight-week-old female Wistar rats were randomly allotted into four groups (n = 5). Administration of letrozole (1 mg/kg p.o) for 21 days induced PCOS, thereafter the animals were treated with 3x109 CFU (p. o) of probiotics for six weeks. RESULTS Letrozole-induced PCOS rats were characterized with elevated circulating testosterone, and multiple ovarian cysts. In addition, rats with PCOS developed increased body weight, which was also accompanied with insulin resistance, hyperinsulinemia, and increased leptin, and decreased adiponectin and adipose TG, as well as elevated adipose lipase. Inflammatory markers (NF-kB, TNF-α) were elevated, while antioxidant defense (GSH) was depleted in PCOS animals. A significant increase in adipose TGF-β1, caspase-6 and HDAC2 levels was observed in PCOS rats when compared with the control. Immunohistochemical evaluation of adipose tissue also showed severe expression of NLRP3 in PCOS rats and these changes were accompanied by increased level of TGF-β1. However, treatment with probiotics reversed these aberrant systemic and adipose tissue changes in PCOS model. CONCLUSION The present results suggest the therapeutic benefit of probiotics against metabolic disturbance in PCOS model through suppression of TGF-β1-dependent pathway.
Collapse
Affiliation(s)
- Kehinde S Olaniyi
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado Ekiti 360101, Nigeria.
| | - Doris O Okara
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado Ekiti 360101, Nigeria
| | - Stephanie E Areloegbe
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado Ekiti 360101, Nigeria
| |
Collapse
|
7
|
Yazdanpanah Z, Cheraghi E, Nasrabadi MH, Salehipour M. Improvement in biochemical manifestations of the serum and follicular fluid and ICSI outcomes in PCOS patients with myo-inositol administration: prospective randomized research. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7187-7198. [PMID: 39718613 DOI: 10.1007/s00210-024-03745-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
The research investigated the capacity of myo-inositol (MI) in order that it improves biochemical markers in serum and follicular fluid and, ultimately, intracytoplasmic sperm injection (ICSI) outcomes of women with PCOS. Sixty infertile patients with PCOS, who were undergoing ovulation induction for ICSI, were randomly divided to two groups. The MI group received 2000 mg myo-inositol + 1 mg folic acid twice a day for 6 weeks with starting the ICSI cycle. For the same period, the control group received a placebo containing only folic acid (1 mg). Levels of hormonal profiles in serum and follicular fluid, as well as oxidative stress markers (MDA, TAC, GPx, and SOD), were estimated using an ELISA assay. Primary end points were ICSI cycle outcomes. Compared to the placebo group, the MI group demonstrated significant reduction in serum and follicular fluid levels of LH, LH/FSH ratio, total testosterone, AMH, and androstenedione. Furthermore, the MI group exhibited meaningful increases in TAC, GPx, and SOD, but MDA significantly decreased. While the number of retrieved and mature oocytes is not statistically similar among the groups, the MI group showed significant improvements in the percentage of immature oocytes, cleavage rate, and good embryo quality. A meaningful correlation was checked between follicular fluid AMH level and LH, FSH, total testosterone, androstenedione, insulin, MDA, the number of retrieved oocytes, and immature oocytes. Our outcomes indicate that myo-inositol administration in women with PCOS undergoing ART helps to improve their hormonal profiles, and the quality of oocytes and embryos. Registration details: Date: 2022.10.19, Registry: https://irct.behdasht.gov.ir/trial/66005 , and Trial registration: IRCT202220921056008N1.
Collapse
Affiliation(s)
- Zeynab Yazdanpanah
- Department of Biology, Parand Branch, Islamic Azad University, Bahonar Blvd, Tehran, Iran
| | - Ebrahim Cheraghi
- Department of Biology, Faculty of Sciences, University of Qom, Qom, Iran
| | | | - Masoud Salehipour
- Department of Biology, Parand Branch, Islamic Azad University, Bahonar Blvd, Tehran, Iran
| |
Collapse
|
8
|
Waldrop SW, Buenaventura M, Campoverde Reyes KJ, Stanford FC. Disparities in the Diagnosis and Management of Polycystic Ovarian Syndrome in Adolescents. Endocrinol Metab Clin North Am 2025; 54:233-250. [PMID: 40348565 DOI: 10.1016/j.ecl.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Addressing polycystic ovary syndrome health disparities requires increasing provider index of suspicion; eliminating implicit bias in diagnosis; making specialty level care accessible to all; establishing a framework of multidisciplinary management and multisectoral care provision that emphasizes longitudinal sustainable lifestyle modifications; and educating and empowering the patient.
Collapse
Affiliation(s)
- Stephanie W Waldrop
- Division of Clinical Sciences, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA; Section on Nutrition, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Nutrition Obesity Research Center (NORC) - University of Colorado, Aurora, CO, USA.
| | - Michaela Buenaventura
- Section on Pediatric Endocrinology, Department of Pediatrics, Children's Hospital of Colorado, University of Colorado Anschutz Medical Campus, 13123 East 16th Avenue Box B265, Aurora, CO 80045, USA
| | - Karen J Campoverde Reyes
- Pediatric Residency Program, Yale School of Medicine, Department of Pediatrics, 330 Cedar Street, LMP 5039, New Haven, CT 06520, USA
| | - Fatima Cody Stanford
- Obesity Medicine, Department of Internal Medicine and Pediatrics, MGH Weight Center, Massachusetts General Hospital; Division of Endocrinology, Department of Medicine, Nutrition Obesity Research Center at Harvard (NORCH), 50 Staniford Street, Suite 430, Boston, MA 02114, USA; Division of Neuroendocrine, Department of Medicine, Nutrition Obesity Research Center at Harvard (NORCH), Boston, MA, USA; Division of Endocrinology, Department of Pediatrics, Nutrition Obesity Research Center at Harvard (NORCH), Boston, MA, USA
| |
Collapse
|
9
|
Carriero VC, Forte G, Dinicola S, Oliva MM, Mudarris GA, Unfer V. Insights from the EGOI-PCOS patient survey: Diagnosis, treatment, and quality of life according to Italian PCOS patients. Eur J Obstet Gynecol Reprod Biol 2025; 310:113947. [PMID: 40179473 DOI: 10.1016/j.ejogrb.2025.113947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/03/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is an endocrine-metabolic disorder; however, the current guidelines do not adequately address the metabolic aspect. By gathering patients' perspectives, this survey investigates potential issues with the current diagnostic process to identify key points that need addressing in the future. METHOD A survey comprising of 49 multiple-choice question was distributed to members of the Italian PCOS community NoiPCOS, including topics such as demographics, PCOS diagnosis experience, symptom management, quality of life, and access to information about PCOS. RESULTS 769 women aged 18-40 responded to the survey. 72.2% of responders were employed and perceived their socio-economic status as "good". PCOS diagnosis was primarily obtained in adolescence (35.1%) or late adolescence (33.6 %), with the most common symptoms being polycystic ovaries (85.8%), irregular menses (80.4%), and hirsutism (64.1%). Moreover, PCOS symptoms were seen to severely impact the mental health for 64.7% of responders. Treatments prescribed for PCOS were diet (49.5%), exercise (46.9%), metformin (27.6%), hormonal contraception (26.4%), and myo-inositol and D-chiro-inositol (25.2%). When accessing information about PCOS, women often relied on unofficial sources (i.e. internet sources) rather guidance from their physician. CONCLUSION Findings of this survey highlight that a thorough update of PCOS diagnostic criteria is required, which should consider the endocrine and metabolic aspects of the syndrome. Such revision should enable a more accurate, precise diagnosis that translates to effective therapy. Finally, any reconsideration of the PCOS guidelines should increase the perceived reliability by patients of medical care, reducing the communication gap between specialists and patients.
Collapse
Affiliation(s)
| | | | - Simona Dinicola
- The Experts Group On Inositol in Basic and Clinical Research and On PCOS (EGOI-PCOS), Rome, Italy; Systems Biology Group Lab, Sapienza University of Rome, Rome, Italy
| | - Mario Montanino Oliva
- The Experts Group On Inositol in Basic and Clinical Research and On PCOS (EGOI-PCOS), Rome, Italy; Department of Obstetrics and Gynecology, Santo Spirito Hospital, Rome, Italy
| | | | - Vittorio Unfer
- The Experts Group On Inositol in Basic and Clinical Research and On PCOS (EGOI-PCOS), Rome, Italy; UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy.
| |
Collapse
|
10
|
Kennard A, Gillespie E, McKendree R. Integrative Medicine in Women. Prim Care 2025; 52:291-306. [PMID: 40412907 DOI: 10.1016/j.pop.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Integrative medicine is a whole-person, holistic approach that combines evidence-based complementary modalities, lifestyle approaches, and Western medicine to comprehensively treat patients. Women are the highest users of integrative modalities in the United States, and women's health conditions commonly benefit from an integrative approach. This study presents evidence-based integrative medicine for common women's health conditions.
Collapse
Affiliation(s)
- Anne Kennard
- Integrative and Lifestyle Medicine, Department of Obstetrics and Gynecology, Marian Regional Medical Center, Santa Maria, CA 93454, USA.
| | - Erin Gillespie
- Integrative Medicine, Internal Medicine, Obesity Medicine, Leap Medical Writing and Editing LLC, Tijeras, NM 87059, USA
| | - Reagan McKendree
- Department of Family Medicine, Marian Regional Medical Center, 1400 E Church Street, Santa Maria, CA 93454, USA
| |
Collapse
|
11
|
Sıgınır A, Bostan H, Saygılı ES, Tufan C, Karakılıc E. Cabergoline monotherapy in polycystic ovary syndrome patients with elevated prolactin: a viable option? Endocrine 2025:10.1007/s12020-025-04279-8. [PMID: 40399716 DOI: 10.1007/s12020-025-04279-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/04/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE Cabergoline is widely used to treat hyperprolactinemia, but its effects on polycystic ovary syndrome (PCOS) remain unclear. Since hyperprolactinemia is present in nearly 30% of PCOS cases, this study aims to assess the impact of cabergoline on androgen levels and clinical outcomes in PCOS with elevated prolactin cases, discussing these findings with the results in prolactinoma cases. METHODS A total of 66 women aged 18-40 were included in this retrospective cohort study, with 36 in the PCOS with elevated prolactin group (median 24.0 (22.0-27.5) years) and 30 in the prolactinoma group (median 28.0 (23.7-33.0) years). Only patients who had been started on cabergoline treatment and had available follow-up data were included. Hormonal profiles and clinical findings, including hirsutism, and menstrual cycle regularity, were assessed before and after cabergoline treatment. RESULTS After cabergoline treatment, significant reductions in prolactin and total testosterone levels were observed in both groups. In the PCOS group, total testosterone decreased from 0.65-0.49 ng/mL (p < 0.001) and dehydroepiandrosterone-sulphate levels from 407.5-301.0 µg/dL (p < 0.001). In the prolactinoma group, total testosterone decreased from 0.39-0.29 ng/mL (p < 0.001). Menstrual irregularities improved markedly in both groups, with prevalence decreasing from 83.3-5.6% in PCOS group and from 80.0-10.0% in the prolactinoma group (p < 0.001). Furthermore, in PCOS group, the prevalence of hirsutism was decreased from 86.1-61.1% (p = 0.007). CONCLUSION Cabergoline is effective in lowering prolactin and androgen levels while improving menstrual regularity in both PCOS and prolactinoma patients, highlighting its potential as a valuable therapeutic option for patients with PCOS with elevated prolactin.
Collapse
Affiliation(s)
- Aslı Sıgınır
- Department of Internal Medicine, Canakkale Onsekiz Mart University Faculty of Medicine, Canakkale, Türkiye
| | - Hayri Bostan
- Department of Endocrinology and Metabolism, Canakkale Mehmet Akif Ersoy State Hospital, Canakkale, Türkiye
| | - Emre Sedar Saygılı
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Canakkale Onsekiz Mart University Faculty of Medicine, Canakkale, Türkiye
| | - Ceren Tufan
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Canakkale Onsekiz Mart University Faculty of Medicine, Canakkale, Türkiye
| | - Ersen Karakılıc
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Canakkale Onsekiz Mart University Faculty of Medicine, Canakkale, Türkiye.
| |
Collapse
|
12
|
Ju S, Kang ZY, Yang LY, Xia YJ, Guo YM, Li S, Yan H, Qi MK, Wang HP, Zhong L. Gut microbiota and ovarian diseases: a new therapeutic perspective. J Ovarian Res 2025; 18:105. [PMID: 40399985 PMCID: PMC12093725 DOI: 10.1186/s13048-025-01684-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/28/2025] [Indexed: 05/23/2025] Open
Abstract
The gut microbiota is a complex community of microorganisms that inhabit the human gastrointestinal tract, helping to maintain the ecological balance of the body's internal and external environments. Disruptions in the composition and diversity of gut microbiota, as well as changes in their metabolic functions, can link to the development and severity of conditions such as premature ovarian insufficiency, polycystic ovary syndrome, and ovarian tumors. This article thoroughly reviews recent research on the connection between gut microbiota and ovarian diseases, providing fresh perspectives on their prevention, pathogenesis, and treatment.
Collapse
Affiliation(s)
- Shan Ju
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Zhen Yang Kang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
- NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China
| | - Li Ya Yang
- NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China
| | - Yong Jun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Yi Ming Guo
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
- NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China
| | - Sui Li
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Hongli Yan
- Changhai hospital, the Navy medical university, 200433, shanghai, China
| | - Ming Kang Qi
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
- NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China
| | - Hui Ping Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China.
| | - Lian Zhong
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, 200093, PR China.
| |
Collapse
|
13
|
Lentini G, Querqui A, Monti N, Bizzarri M. PCOS and Inositols - Advances and Lessons We are Learning. A Narrative Review. Drug Des Devel Ther 2025; 19:4183-4199. [PMID: 40420946 PMCID: PMC12104671 DOI: 10.2147/dddt.s524718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction This Expert Opinion covers recent updates in the use of Inositol in polycystic ovary syndrome (PCOS), highlighting the specific effects triggered upon ovarian steroidogenesis. Areas Covered An impressive body of evidence, obtained from molecular, animal and clinical studies, demonstrated the striking association between PCOS and the metabolism of myo-Inositol (myo-Ins) and its isomer D-Chiro-Inositol (DCI). Early investigations focused primarily on the metabolic consequences of inositol in modulating insulin transduction. However, recent advances disclosed that Inositols trigger direct effects on steroidogenesis. High DCI levels exacerbate androgen synthesis, and downregulate aromatase expression. Myo-Ins modulates insulin effects too, but exerts opposite actions on steroidogenesis, by increasing aromatase and FSH receptor expression. Clinical studies demonstrated myo-Ins efficacy, suggesting that an appropriate ratio in between myo-Ins/DCI (40:1) improves the reproductive function in PCOS women, even in absence of insulin resistance. Expert Opinion Inositol-based treatments in PCOS are gaining momentum, demonstrating safety and efficacy greater than those obtained with other pharmacological agents. The efficacy depends not only on the modulation of insulin sensitivity but also on the direct, steroidogenic effects upon the ovaries. Adequate adsorption of Inositol is a critical issue, and the association of α-Lactalbumin can significantly overcome this problem. However, if a treatment based on inositol could be equally effective on different phenotypes of PCOS needs a specific assessment.
Collapse
Affiliation(s)
- Guglielmo Lentini
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Alessandro Querqui
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Noemi Monti
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Space Biomedicine Laboratory, University Sapienza, Rome, Italy
| |
Collapse
|
14
|
Basnet J, Rezq S, Huffman AM, Asala TE, Yanes Cardozo LL, Romero DG. Androgen Receptor PROTAC ARV-110 Ameliorates Metabolic Complications in a Mouse Model of Polycystic Ovary Syndrome. Endocrinology 2025; 166:bqaf091. [PMID: 40437805 PMCID: PMC12120138 DOI: 10.1210/endocr/bqaf091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Indexed: 06/01/2025]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive-age women. Hyperandrogenemia (HA) is a hallmark of PCOS and is positively associated with metabolic complications. Androgens exert their biological actions through the androgen receptor (AR), which regulates transcriptional activity. Antiandrogens are not recommended for managing metabolic complications in PCOS due to their hepatotoxicity, despite being a viable therapy to treat HA. We hypothesized that the novel AR Proteolysis Targeting Chimera (PROTAC) degrader ARV-110 would downregulate AR protein levels and actions to abolish or mitigate HA-mediated metabolic complications using a well-established HA mouse model of PCOS. Three-week-old female mice were implanted with dihydrotestosterone (DHT) or control pellets. Four weeks later, mice were treated with low- (ARV-110-L, 1 mg/kg.day) or high-dose (ARV-110-H, 10 mg/kg.day) ARV-110 for an additional 8 weeks. ARV-110 dose-dependently reduced AR protein levels in white adipose tissue (WAT), kidney, liver, and ovary. ARV-110 attenuated DHT-induced increases in body weight, fat mass, kidney mass, WAT mass, circulating leptin and antimüllerian hormone, and altered glucose homeostasis. ARV-110-H increased kidney (UACR, KIM-1, NGAL) and liver (ALT, AST, LDH) injury markers and caused severe hepatomegaly, while ARV-110-L mostly spared those deleterious effects. Unbiased proteomics analysis revealed that ARV-110-H treatment severely affected the liver proteome and dysregulated multiple signaling and metabolic canonical pathways, while only minimal effects were observed with ARV-110-L treatment. In summary, our findings underscore the potential of AR PROTACs as a novel therapeutic approach for managing metabolic complications in PCOS, provided the dosing is carefully optimized to avoid adverse effects.
Collapse
Affiliation(s)
- Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alexandra M Huffman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Tolulope E Asala
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine (Endocrinology), University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
15
|
Marchesan LB, da Silva TR, Spritzer PM. Topiramate Added to Metformin for Obesity Control in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2025; 110:e1892-e1901. [PMID: 39271474 DOI: 10.1210/clinem/dgae637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is often linked with obesity, and weight management can improve endocrine and cardiometabolic features. OBJECTIVE This work aimed to evaluate the effects of adding topiramate (TPM) to metformin (MTF) on weight control and hormonal and metabolic outcomes in women with PCOS. METHODS In a randomized, double-blind, placebo-controlled trial, participants with PCOS and body mass index of 30 or greater, or 27 or greater associated with hypertension, type 2 diabetes, or dyslipidemia followed a 20 kcal/kg diet in addition to 850 mg of MTF or a previous MTF regimen. They were randomly assigned to receive either TPM or placebo (P) alongside MTF. Anthropometric measurements, blood pressure, modified Ferriman-Gallwey score (mFGS), and adverse events were assessed every 4 weeks for 6 months. The primary end point was the percentage change in body weight from baseline in both groups. Secondary end points included changes in clinical, cardiometabolic, and hormonal parameters and psychosocial features. RESULTS Thirty-one participants were in the MTF + P group and 30 in the MTF + TPM group. The MTF + TPM group showed greater mean weight loss at 3 months (-3.4% vs -1.6%; P = .03) and 6 months (-4.5% vs -1.4%; P = .03). Both groups had improved androgens, lipids, and psychosocial scores. Participants with 3% or greater weight loss at 6 months had improved mFGS (8.4 to 6.5; P = .026). Paresthesia was more common in the MTF + TPM group (23.3% vs 3.2%; P = .026). CONCLUSION Combining TPM with MTF and a low-calorie diet may be an effective, low-cost, easy-to-use, and safe strategy for weight management in women with PCOS, with mild adverse effects.
Collapse
Affiliation(s)
- Lucas Bandeira Marchesan
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
- Division of Endocrinology, Hospital Nossa Senhora da Conceição (HNSC), Porto Alegre, RS 91350-200, Brazil
- Postgraduate Program in Endocrinology, Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Thais Rasia da Silva
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
- Postgraduate Program in Endocrinology, Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Poli Mara Spritzer
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
- Postgraduate Program in Endocrinology, Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
- Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| |
Collapse
|
16
|
Hu S, Lalonde-Bester S, Salem J, Koshy S, Vine D, Harrison TG, Yamamoto JM, Benham JL. Validity of administrative health data case definitions for identifying polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod 2025:deaf094. [PMID: 40381996 DOI: 10.1093/humrep/deaf094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/17/2025] [Indexed: 05/20/2025] Open
Abstract
STUDY QUESTION What is the validity of published administrative health data case definitions of polycystic ovary syndrome (PCOS) compared with reference standards? SUMMARY ANSWER Due to the limited number of eligible studies, drawing definitive conclusions is challenging; however, this review highlights significant gaps and variability in current PCOS case definitions, underscoring the need for standardized case definitions in future research. WHAT IS KNOWN ALREADY Administrative health data offer the opportunity to evaluate health outcomes and disease epidemiology at a population-level. Currently, the validity of existing administrative health data case definitions for PCOS is unknown. STUDY DESIGN, SIZE, DURATION A systematic review of the literature was conducted on full-text English-language articles up to July 2023, using the MEDLINE and EMBASE databases. PARTICIPANTS/MATERIALS, SETTING, METHODS Two reviewers independently screened titles, abstracts and full texts, extracted data, assessed study quality and graded validity. A random effects meta-analysis was conducted to pool reported validity measures and heterogeneity was examined. MAIN RESULTS AND THE ROLE OF CHANCE The review included four eligible articles consisting of three cross-sectional studies and one retrospective cohort study. Two studies defined PCOS using the Rotterdam Criteria, one study used self-report, and one used a clinical gold standard. All case definitions included the International Classification of Diseases (ICD)-9 code 256.4 for 'polycystic ovaries' and three studies used E28.2 for 'polycystic ovarian syndrome'. Three studies reported positive predictive value (PPV), which ranged from 30 to 96%. One study reported both PPV (96%) and sensitivity (50%) for one case definition. The pooled PPV estimate for the ICD code-based case definitions was 88% (95% confidence interval 82-95%; I2 = 100%). One study reported fair agreement (percent agreement= 90.3, κ = 0.27, percent agreement bias adjusted κ = 0.81). Overall, the risk of bias of the included studies was low. LIMITATIONS, REASONS FOR CAUTION There were limited number of validations and precision indices of validations. WIDER IMPLICATIONS OF THE FINDINGS Further validation of these case definitions in other administrative health datasets, and development of novel coding algorithms is required to inform future population-based studies in PCOS. STUDY FUNDING/COMPETING INTEREST(S) No external funding was used and there are no disclosures. REGISTRATION NUMBER PROSPERO CRD42023385617.
Collapse
Affiliation(s)
- Sophie Hu
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | - Jenna Salem
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sheffinea Koshy
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Donna Vine
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tyrone G Harrison
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- O'Brien Institute for Public Health and Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Jennifer M Yamamoto
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Children's Hospital Research Institute of Manitoba, Children's Hospital Foundation of Manitoba, Winnipeg, MB, Canada
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jamie L Benham
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- O'Brien Institute for Public Health and Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
17
|
Vale-Fernandes E, Carrageta DF, Moreira MV, Guerra-Carvalho B, Rodrigues B, Sousa D, Brandão R, Leal C, Barreiro M, Tomé A, Alves MG, Oliveira PF, Monteiro MP. Follicular fluid profiling unveils anti-Müllerian hormone alongside glycolytic and mitochondrial dysfunction as markers of polycystic ovary syndrome. Mol Cell Endocrinol 2025; 602:112536. [PMID: 40185328 DOI: 10.1016/j.mce.2025.112536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, yet the molecular mechanisms influencing its pathophysiology remain poorly defined. A comprehensive prospective case-control study was conducted to elucidate the follicular fluid (FF) hormone and metabolite profile in women with PCOS and its implications for oocyte maturation and fertilization. The study involved 40 age- and body mass index (BMI)-matched women undergoing in vitro fertilization (IVF), including 20 diagnosed with PCOS and 20 controls with infertility due to tubal or male factors. A distinctive hormone profile in the FF of women with PCOS was identified, characterized by significantly higher anti-Müllerian hormone (AMH) levels (24.90 ± 17.61 vs. 16.68 ± 17.67 pmol/L, p = 0.0039) and lower progesterone (8253 ± 4748 vs. 25362 ± 10862 ng/mL, p < 0.0001) and estradiol levels (388.23 ± 210.58 vs. 651.48 ± 390.79 ng/mL, p = 0.0208) compared to normoovulatory controls. Moreover, a metabolite fingerprint associated with glycolytic and mitochondrial dysfunction was observed, as evidenced by lower lactate (4575.44 ± 1507.76 vs. 5595.34 ± 1073.32 μmol/L, p = 0.0182) and formate (64.51 ± 16.06 vs. 75.81 ± 16.63 μmol/L, p = 0.0351) levels and higher citrate levels (136.93 ± 52.53 vs. 109.15 ± 24.17 μmol/L, p = 0.0409) in the FF of women with PCOS. These findings suggest that the molecular profile of the FF in women with PCOS might be related to granulosa cell glycolytic and mitochondrial dysfunction, which can have a negative impact on oocyte fertilization potential. The study provides an integrative analysis of the FF hormone and metabolite profile in women with PCOS, offering insights into the molecular mechanisms underlying the reproductive dysfunctions associated with this condition.
Collapse
Affiliation(s)
- Emídio Vale-Fernandes
- Centre for Medically Assisted Procreation/Public Gamete Bank, Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal; Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal; Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal.
| | - David F Carrageta
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal; Portalegre Polytechnic Institute, 7300-110, Portalegre, Portugal
| | - Mafalda V Moreira
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal
| | - Bárbara Guerra-Carvalho
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal
| | - Bárbara Rodrigues
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; Molecular Genetics Laboratory, Laboratory Genetics Service, Genetics and Pathology Clinic, Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal
| | - Daniela Sousa
- Centre for Medically Assisted Procreation/Public Gamete Bank, Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal
| | - Raquel Brandão
- Centre for Medically Assisted Procreation/Public Gamete Bank, Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal
| | - Carla Leal
- Centre for Medically Assisted Procreation/Public Gamete Bank, Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal; Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal
| | - Márcia Barreiro
- Centre for Medically Assisted Procreation/Public Gamete Bank, Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal; Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal; Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal
| | - António Tomé
- Gynecology Department, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário de Santo António (CHUdSA), Unidade Local de Saúde de Santo António (ULSSA), 4099-001, Porto, Portugal
| | - Marco G Alves
- iBiMED - Institute of Biomedicine and Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal
| |
Collapse
|
18
|
Reka S, Praba TS, Prasanna M, Reddy VNN, Amirtharajan R. Automated high precision PCOS detection through a segment anything model on super resolution ultrasound ovary images. Sci Rep 2025; 15:16832. [PMID: 40369044 PMCID: PMC12078606 DOI: 10.1038/s41598-025-01744-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 05/08/2025] [Indexed: 05/16/2025] Open
Abstract
PCOS (Poly-Cystic Ovary Syndrome) is a multifaceted disorder that often affects the ovarian morphology of women of their reproductive age, resulting in the development of numerous cysts on the ovaries. Ultrasound imaging typically diagnoses PCOS, which helps clinicians assess the size, shape, and existence of cysts in the ovaries. Nevertheless, manual ultrasound image analysis is often challenging and time-consuming, resulting in inter-observer variability. To effectively treat PCOS and prevent its long-term effects, prompt and accurate diagnosis is crucial. In such cases, a prediction model based on deep learning can help physicians by streamlining the diagnosis procedure, reducing time and potential errors. This article proposes a novel integrated approach, QEI-SAM (Quality Enhanced Image - Segment Anything Model), for enhancing image quality and ovarian cyst segmentation for accurate prediction. GAN (Generative Adversarial Networks) and CNN (Convolutional Neural Networks) are the most recent cutting-edge innovations that have supported the system in attaining the expected result. The proposed QEI-SAM model used Enhanced Super Resolution Generative Adversarial Networks (ESRGAN) for image enhancement to increase the resolution, sharpening the edges and restoring the finer structure of the ultrasound ovary images and achieved a better SSIM of 0.938, PSNR value of 38.60 and LPIPS value of 0.0859. Then, it incorporates the Segment Anything Model (SAM) to segment ovarian cysts and achieve the highest Dice coefficient of 0.9501 and IoU score of 0.9050. Furthermore, Convolutional Neural Network - ResNet 50, ResNet 101, VGG 16, VGG 19, AlexNet and Inception v3 have been implemented to diagnose PCOS promptly. Finally, VGG 19 has achieved the highest accuracy of 99.31%.
Collapse
Affiliation(s)
- S Reka
- School of Computing, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, India
| | - T Suriya Praba
- School of Computing, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, India.
| | - Mukesh Prasanna
- School of Computing, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, India
| | | | - Rengarajan Amirtharajan
- School of Electrical and Electronics Engineering, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, India
| |
Collapse
|
19
|
Firat S, Elter K, Ateş S, Fisunoğlu M. Does MYO and ALA Supplementation Improve PCOS Outcomes? MEDICINA (KAUNAS, LITHUANIA) 2025; 61:885. [PMID: 40428843 PMCID: PMC12113115 DOI: 10.3390/medicina61050885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/07/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: This study aimed to evaluate the impact of myoinositol (MYO) and α-lipoic acid (ALA) supplementation on hormonal and metabolic markers in women diagnosed with polycystic ovary syndrome (PCOS). Materials and Methods: A retrospective case-control study was conducted with 58 women aged between 18-40 years who met the Rotterdam criteria for PCOS. The case group (n = 29) received MYO (2000 mg/day) and ALA (400 mg/day) supplements, while the control group (n = 29) did not receive any treatment. Data on the subjects' anthropometric measures, glycemic indices, sex hormones, and lipid profiles were collected. Results: The results demonstrated that, following three months of MYO + ALA supplementation, the case group exhibited steady body weight (p = 0.484) and BMI (p = 0.405), whereas the control group demonstrated a significant increase in both (p = 0.029; p = 0.026, respectively). A stratified analysis based on BMI, waist circumference, and waist-to-height ratio revealed that HbA1c (%) was significantly lower in the "normal" subgroup compared to the "risky" subgroup within the case group (p < 0.05). Although the mean HbA1c, insulin, and HOMA-IR values were comparable between the two groups, the LH/FSH ratio significantly increased in the control group (p = 0.010). No significant differences were observed in the lipid profiles between the two groups; however, LDL levels decreased significantly in the case group (p = 0.024). Across all classifications, the "normal" subgroup consistently exhibited lower HbA1c and TG/HDL ratios than the "risky" subgroup. Conclusions: Adding MYO + ALA supplementation to standard PCOS treatment may offer metabolic benefits, particularly in maintaining glycemic control, body weight, and BMI. Supplementation also reduces LDL.
Collapse
Affiliation(s)
- Selma Firat
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Kırklareli University, Kırklareli 39100, Turkey
| | - Koray Elter
- Department of Obstetrics and Gynecology, Faculty of Medicine, Trakya University, Edirne 22030, Turkey or (K.E.); (S.A.)
| | - Sinan Ateş
- Department of Obstetrics and Gynecology, Faculty of Medicine, Trakya University, Edirne 22030, Turkey or (K.E.); (S.A.)
| | - Mehmet Fisunoğlu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06100, Turkey;
| |
Collapse
|
20
|
Carmina E. The Role of Gene Alterations in the Pathogenesis of Polycystic Ovary Syndrome. J Clin Med 2025; 14:3347. [PMID: 40429342 PMCID: PMC12112259 DOI: 10.3390/jcm14103347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/20/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Family studies have shown that polycystic ovary syndrome (PCOS) has probable genetic transmission because of a high incidence of relatives who present clinical or biochemical characters of the syndrome. However, initial candidate gene studies were unsuccessful. Genome wide association studies (GWASs) have shown that at least 29 gene alterations are common in PCOS, but it has been calculated that the altered genes found by GWASs may represent only 10% of affected patients. Rare altered uncoding genes may explain the syndrome in an additional group of patients. In many other patients, the altered genes found by GWASs may represent a risk condition for the development of the syndrome, and new candidate gene studies have shown that some gene alterations that mainly concern androgen production may be common in PCOS. Finally, in most patients, epigenetic and environmental factors may be necessary to transform a risk condition into this common and important syndrome.
Collapse
Affiliation(s)
- Enrico Carmina
- Endocrinology Unit, School of Medicine, University of Palermo, 90139 Palermo, Italy
| |
Collapse
|
21
|
Jafari K, Tajik N, Moini A, SeyedAlinaghi S, Abiri A. Metabolic mediators of the overweight's effect on infertility in women with polycystic ovary syndrome. Sci Rep 2025; 15:16258. [PMID: 40346143 PMCID: PMC12064820 DOI: 10.1038/s41598-025-01287-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
Women with polycystic ovary syndrome (PCOS) often experience infertility, potentially mediated by metabolic factors altered by elevated body mass index (BMI). While triglycerides (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and fasting blood sugar (FBS) are known mediators in the BMI-infertility relationship, the extent of their mediation effects remains unquantified in prior studies. This study quantifies the mediation effect of these metabolic factors. We conducted a cross-sectional study on 669 women diagnosed with PCOS at a tertiary hospital in Tehran, Iran, from 2021 to 2023. Data on BMI, TG, LDL, HDL, and FBS were collected, with infertility defined as the inability to conceive after 12 months of unprotected intercourse. Mediation analysis was performed using the Karlson Holm Breen (KHB) method, adjusting for age. Higher BMI was associated with increased levels of TG, LDL, and FBS and decreased HDL, all of which (except for LDL) were linked to infertility. Mediation analysis revealed that after adjusting for age, TG, HDL, and FBS significantly mediated the BMI-infertility association, accounting for 52.98%, 79.19%, and 49.7% of the effect, respectively. Our study identified TG, HDL, and FBS as significant mediators of the BMI-infertility link, with over half of the association mediated through these factors. Targeting metabolic improvements may help reduce infertility risk in this population.
Collapse
Affiliation(s)
- Kasra Jafari
- Research Development Center, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Epidemiology, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Nooshan Tajik
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Moini
- Department of Obstetrics and Gynecology, Endocrinology and Female Infertility Unit, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Breast Disease Research Center (BDRC), Tehran University Of Medical Sciences, Tehran, Iran
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - SeyedAhmad SeyedAlinaghi
- Research Development Center, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Institute for Reduction of High-Risk Behaviors, Iranian Research Center for HIV/AIDS, Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Abiri
- Department of Obstetrics and Gynecology, School of Medicine, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Obstetrics and Gynecology, School of Medicine, Arash Women's Hospital, Tehran University of Medical Sciences, Eastern 162th St., Baghdarnia St., Resalat Highway, Tehranpars, PO box: 1653915981, Tehran, Iran.
| |
Collapse
|
22
|
Luyckx L, Myllykangas M, Saarela U, Virtanen N, Hurskainen E, Savolainen A, Ollikainen N, Norlén AK, Ohlsson C, Poutanen M, Velde GV, Arffman RK, Prunskaite-Hyyryläinen R, Vriens J, Piltonen TT. Prenatally androgenized PCOS mice have ovary-independent uterine dysfunction and placental inflammation aggravated by high-fat diet. SCIENCE ADVANCES 2025; 11:eadu3699. [PMID: 40344073 PMCID: PMC12063661 DOI: 10.1126/sciadv.adu3699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a common hyperandrogenic and metabolic condition in women. The syndrome is linked to subfertility and pregnancy complications, yet the independent effects of exposure to hyperandrogenism and obesity on endometrial function remain unclear. Here, PCOS-like mice were generated using prenatal androgenization (PNA) with dihydrotestosterone, followed by a prepubertal high-fat (HF) or standard diet. In ovariectomized mice, PNA impaired uterine closure during the implantation window, disrupted decidualization, and altered extracellular matrix- and inflammation-related gene expression. The effects were aggravated by the HF diet. In naturally mated, ovary-intact mice, PNA and HF diet affected decidual and placental gene expression, suggestive of placental dysfunction and inflammation, and induced fetal growth restriction. This study underlines the role of the uterus in adverse pregnancy outcomes in PCOS and identifies possible underlying mechanisms for future studies. Prepregnancy interventions targeting metabolic health and hyperandrogenism should be the next steps to optimize PCOS pregnancy outcomes.
Collapse
Affiliation(s)
- Lena Luyckx
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Research Group for Implantation, Placentation and Pregnancy, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Milena Myllykangas
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nikke Virtanen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Elisa Hurskainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Audrey Savolainen
- Protein and Structural Biology Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nadja Ollikainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Anna-Karin Norlén
- Department of Clinical Chemistry, Faculty of Medicine, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, 41345 Gothenburg, Sweden
| | - Matti Poutanen
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, 41345 Gothenburg, Sweden
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Greetje Vande Velde
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Riikka K. Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Renata Prunskaite-Hyyryläinen
- Protein and Structural Biology Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland
| | - Joris Vriens
- Research Group for Implantation, Placentation and Pregnancy, Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| |
Collapse
|
23
|
Sarray S, Ezzidi I, Ben Salem A, Ben Abdennebi H, Mtiraoui N. The Role of AMH and AMHR2 Variants in Polycystic Ovary Syndrome: 'A Comprehensive Analysis'. Clin Endocrinol (Oxf) 2025. [PMID: 40344233 DOI: 10.1111/cen.15269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/28/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Variants in the genes encoding anti-mullerian hormone (AMH) and its receptor 2 (AMHR2) have been identified as potential contributors to the development of polycystic ovary syndrome (PCOS). However, results from association studies examining their role in PCOS have been inconsistent. This study aims to investigate the potential association between AMH and AMHR2 gene variants and the risk of PCOS as well as their influence on serum AMH levels in a Tunisian cohort. METHODS The case-control study recruited 327 PCOS women and 396 healthy controls. DNA was extracted and genotyped for three variants in the AMH gene namely, rs4807216, rs10407022 and rs8112524 as well as three variants in the AMHR2 gene, including rs2002555, rs11170553 and rs2272002, using the TaqMan SNP genotyping assay. Fasting serum AMH levels were quantified using ELISA. RESULTS Significant metabolic differences were observed in the PCOS cohort, including higher BMI, and elevated levels of AMH, glucose, triglycerides, and cholesterol, along with lower FSH levels. The investigation of genetic associations between AMH and AMHR2 gene variants and PCOS susceptibility revealed notable genotype-specific correlations with lipid profiles. Specifically, the AMH rs8112524 A/A and G/A genotypes were correlated to increased triglyceride levels, while the AMHR2 rs2002555 G/G genotype, as well as the rs11170553 T/T and C/T genotypes, were correlated with decreased HDL levels. However, no significant allelic, genotypic or haplotypic associations were identified, nor was any substantial impact on serum AMH levels observed. Additionally, interaction and epistasis analyses indicated that the AMH and AMHR2 variants had no significant predictive capabilities regarding PCOS susceptibility. CONCLUSION Although AMH and AMHR2 variants may not directly influence PCOS susceptibility, they could play a role in modulating lipid metabolism associated with the syndrome.
Collapse
Affiliation(s)
- Sameh Sarray
- College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
- Faculty of Sciences, University of Tunis EL Manar, Tunis, Tunisia
| | - Intissar Ezzidi
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Assila Ben Salem
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
| | - Hassen Ben Abdennebi
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
| | - Nabil Mtiraoui
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
24
|
Amirkhizi F, Taghizadeh M, Hamedi-Shahraki S, Asghari S. Association of dietary phytochemical index with metabolic markers, serum asymmetric dimethylarginine and atherogenic indices in patients with polycystic ovary syndrome. Nutr Metab (Lond) 2025; 22:39. [PMID: 40336098 PMCID: PMC12060492 DOI: 10.1186/s12986-025-00932-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is associated with an increased risk of cardiovascular diseases (CVD). Plant-based diets are associated with reduced CVD risk factors. This study aimed to explore the associations between dietary phytochemical index (DPI) and asymmetric dimethylarginine (ADMA), lipid profile, atherogenic indices, and other metabolic biomarkers in women with PCOS. METHODS In this cross-sectional study, 150 females aged 18-45 years diagnosed with PCOS were recruited. An interviewer-administered questionnaire was applied to gather the relevant demographic characteristics, detailed clinical information, and lifestyle habits of participants. A validated semi-quantitative food frequency questionnaire was used to assess dietary intake, and DPI was calculated accordingly. We used multiple linear regression to determine the association between serum concentrations of ADMA, total testosterone, sex hormone-binding globulin (SHBG), fasting serum glucose (FSG), insulin, and lipid profile, as well as atherogenic indices across quartiles of DPI. RESULTS There was a negative correlation between the DPI and serum levels of ADMA (p-trend = 0.022), triglycerides (TG) (p-trend = 0.003), oxidized low-density lipoprotein cholesterol (ox-LDL) (p-trend = 0.001), insulin (p-trend = 0.045) and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) (p-trend = 0.018). Moreover, there was a tendency for visceral adiposity index (VAI) (p-trend = 0.005) and atherogenic index of plasma (AIP) (p-trend = 0.001) to decrease as the quartile categories of DPI increased. No significant regular trend was found for serum levels of FSG, SHBG, total testosterone, other lipid profiles, and lipid accumulation product (LAP). CONCLUSIONS These findings suggest that adherence to a phytochemical-rich diet decrease the CVD risk factors in PCOS patients.
Collapse
Affiliation(s)
- Farshad Amirkhizi
- Department of Nutrition, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahdiyeh Taghizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Zabol University of Medical Sciences, Bagheri St., Shahid Rajaei St, Zabol, 9861615881, Iran.
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran.
| |
Collapse
|
25
|
Husni M, Rizk DEE, Alabdulla NS, Zayed A, Malas H, Modahka NA, Alqujan SJ, Alansari BK, Alhajri SE, Mahmood N, Jahrami H. Psychiatric disorders, impulsivity and borderline personality in patients with polycystic ovary syndrome. Arch Womens Ment Health 2025:10.1007/s00737-025-01589-8. [PMID: 40327076 DOI: 10.1007/s00737-025-01589-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION Polycystic ovarian syndrome (PCOS) is a common endocrine condition in women. Studies have shown that PCOS is associated with poor quality of life, anxiety, sadness, dissatisfaction with one's appearance, and sexual dysfunction. OBJECTIVE This study was conducted to determine whether a strong psycho-pathological personality is related to PCOS and whether this personality is related to the hyperandrogenic state. METHODS Anthropometric, metabolic, hormonal, clinical, and psychological characteristics were examined in 90 Bahraini women with PCOS. After confirming the diagnosis of PCOS via Rotterdam criteria, including ovarian ultrasound, each patient was evaluated via the following questionnaires: 1) the GAD-7 (General Anxiety Disorder-7) to measure the severity of anxiety; 2) the Patient Health Questionnaire-9 (PHQ-9) to confirm and measure the severity of depression; 3) the Barratt Impulsiveness Scale (BIS-11) to measure aggression; and 4) the McLean Screening Instrument to identify borderline personality disorders (MSI-BPDs). The study was approved by the Institutional Review Board. RESULTS Compared to controls, PCOS patients exhibited significantly higher rates of severe depression (8% vs. 0%, p < 0.001), severe anxiety (7% vs. 0%, p < 0.001), impulsivity (BIS-11: 39.43 ± 9.69 vs. 26.64 ± 2.92, p < 0.001), and borderline personality traits (McLean: 2.41 ± 2.44 vs. 1.2 ± 0.94, p < 0.001). Metabolic comorbidities, including obesity (BMI 28.88 vs. 20.27, p < 0.001) and hypothyroidism (48% vs. 0%, p < 0.001), were prevalent in PCOS. Hyperandrogenism correlated weakly with psychiatric outcomes (all p > 0.05). CONCLUSION Women with PCOS demonstrate markedly elevated psychiatric and medical burdens compared to healthy controls. While hyperandrogenism showed limited direct associations, metabolic dysfunction (e.g., obesity) may mediate psychiatric risk. These findings underscore the need for multidisciplinary care integrating psychological and endocrine management, particularly in populations where cultural norms exacerbate PCOS-related distress.
Collapse
Affiliation(s)
- Mariwan Husni
- Department of Psychiatry, Northern Ontario School of Medicine, Thunder Bay, ON, Canada.
- Department of Pharmacy, College of Pharmacy, Knowledge University, Erbil, 44001, Iraq.
| | - Diaa E E Rizk
- College of Medicine and Health Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain
| | | | - Aldana Zayed
- King Hamad University, Manama, Kingdom of Bahrain
| | - Hosni Malas
- King Hamad University, Manama, Kingdom of Bahrain
| | | | | | | | | | - Nada Mahmood
- King Hamad University, Manama, Kingdom of Bahrain
| | - Haitham Jahrami
- College of Medicine and Health Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain.
- Government Hospitals, Manama, Kingdom of Bahrain.
| |
Collapse
|
26
|
Maan P, Gautam R, Vasudevan S, Menon GR, Arora A, Nair A, Jabbar PK, Arora T. Pharmacological and Non-Pharmacological Interventions for Polycystic Ovary Syndrome (PCOS) in Indian Women: A Systematic Review and Meta-Analysis. Pharmaceuticals (Basel) 2025; 18:680. [PMID: 40430499 PMCID: PMC12114445 DOI: 10.3390/ph18050680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is an endocrine disorder prevalent in women of reproductive age. Treatment or management of this syndrome includes several pharmacological and non-pharmacological treatment approaches for different manifestations of the disease that vary with the patient's age, symptoms, requirements, and geographical location. Objective: This systematic review aims to conduct a comprehensive and evidence-based analysis of the various available treatment options and identify knowledge gaps in PCOS management in India. Methods: A comprehensive search was conducted in PubMed, Scopus, and Embase databases from January 2010 till February 2024. We included randomized control trials (RCTs) using any pharmacological drugs (e.g., insulin sensitizers, anti-androgens, anti-obesity drugs, oral contraceptive pills, ovulation induction drugs, etc.) or non-pharmacological intervention (e.g., yoga, diet, herbal supplements, etc.) with Indian PCOS patients for improving common manifestations of PCOS and written in the English language. Studies were screened by two authors independently in a two-level process. Data extraction was also performed by two authors. Risk of bias was performed using the RoB 2 Tool. Subgroup analysis and meta-analysis were performed using the RevMan tool. Results: Thirty RCTs on pharmacological and eight on non-pharmacological interventions were included in the study. However, all the RCTs were so heterogeneous in terms of intervention used, subject recruited, and outcomes measured that meta-analysis was possible for only three subgroups (metformin vs. inositol, metformin vs. metformin+ inositol, and letrozole vs. clomiphene citrate), with only two or three studies per analysis. Most studies were single-centric and small-sized and had a high risk of bias, limiting their generalizability. Conclusions: This systematic review synthesized existing research and evaluated the effectiveness and safety of existing treatments. Limitations and gaps in the current research were identified, which may inform future research for better understanding and management of PCOS in the Indian context.
Collapse
Affiliation(s)
- Pratibha Maan
- Indian Council of Medical Research (ICMR) Headquarters, New Delhi 110029, India; (P.M.); (R.G.); (S.V.)
| | - Rohit Gautam
- Indian Council of Medical Research (ICMR) Headquarters, New Delhi 110029, India; (P.M.); (R.G.); (S.V.)
| | - Sudharsan Vasudevan
- Indian Council of Medical Research (ICMR) Headquarters, New Delhi 110029, India; (P.M.); (R.G.); (S.V.)
| | - Geetha R. Menon
- Department of Health Research, Ministry of Health and Family Welfare, Government of India, New Delhi 110001, India;
| | - Amit Arora
- Sir Ganga Ram Hospital, New Delhi 110060, India;
| | - Abilash Nair
- Department of Endocrinology, Government Medical College, Thiruvananthapuram 695011, India
| | | | - Taruna Arora
- Indian Council of Medical Research (ICMR) Headquarters, New Delhi 110029, India; (P.M.); (R.G.); (S.V.)
| |
Collapse
|
27
|
Colonetti L, Uggioni MLR, Prestes GDS, Stangherlin L, Junior JCD, Moura R, Sipriano EDS, Madeira K, Cardoso HS, Ferraz SD, Baptista MM, Grande AJ, Ceretta LB, da Rosa MI, Colonetti T. Effects of carbohydrate reduced diet associated with strength training on clinical signs of women with polycystic ovary syndrome: Randomized clinical trial. Nutrition 2025; 133:112696. [PMID: 40048764 DOI: 10.1016/j.nut.2025.112696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE To evaluate the effects of a low-carbohydrate diet associated with strength training on the clinical signs of polycystic ovary syndrome (PCOS). RESEARCH METHODS AND PROCEDURES A randomized clinical trial was carried out including 29 women over 18 years old diagnosed with PCOS, randomized into two groups, with follow-up for 12 weeks: the low-carbohydrate diet group associated with strength exercise (LCDE); and the standard diet group associated with strength exercise (SDE). We evaluated manifestations of acne, hirsutism by the Ferriman-Gallwey scale and alopecia by the Ludwig-Savin scale, and assessed laboratory tests for total and free testosterone, dehydroepiandrosterone, follicle-stimulating hormone, and luteinizing hormone. The collected data were analyzed using IBM-SPSS software version 21. RESULTS The study showed statistically significant differences in the hormonal levels of dehydroepiandrosterone (P = 0.045), luteinizing hormone (P = 0.017) and follicle-stimulating hormone (P = 0.014) when comparing the LCDE and SDE groups. CONCLUSIONS the intervention used can promote an improvement in the clinical presentation of PCOS, especially in hormonal parameters. The clinical trial was registered on the REBEC platform (Brazilian Registry of Clinical Trials) under number RBR4wjqxcv (Carbohydrate Reduction and Exercise in Women with PCOS) and is available on the website: https://ensaiosclinicos.gov.br/rg/RBR-4wjqxcv.
Collapse
Affiliation(s)
- Laura Colonetti
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Maria Laura Rodrigues Uggioni
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Gabriele da Silveira Prestes
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Luana Stangherlin
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - João Carlos Denoni Junior
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil.
| | - Rafael Moura
- Postgraduate Program in Collective Health, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Erica da Silva Sipriano
- Quantitative Methods Research Group, Laboratory of Applied Research in Computing and Quantitative Methods, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Kristian Madeira
- Quantitative Methods Research Group, Laboratory of Applied Research in Computing and Quantitative Methods, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Hemmylly Silveira Cardoso
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Sarah Dagostin Ferraz
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Mateus Marcelo Baptista
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Antonio Jose Grande
- Laboratory of Evidence-Based Practice, State University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Luciane Bisognin Ceretta
- Postgraduate Program in Collective Health, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Maria Inês da Rosa
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Tamy Colonetti
- Epidemiology Group, Laboratory of Biomedicine Translational, University of the Extreme South of Santa Catarina, Criciúma, Santa Catarina, Brazil
| |
Collapse
|
28
|
Farhan M, Seyfi A, Alnuaimi A, Alamour M, Alwarafi S, Elastal H, Nazir MH, Kamaraj B, Putta Nagarajan HD, Delianne D, Ganesan S, Patel T. A narrative review on cutaneous manifestations in polycystic ovary syndrome: pathophysiology, diagnosis, management, and psychosocial impact. Ann Med Surg (Lond) 2025; 87:2804-2811. [PMID: 40337376 PMCID: PMC12055046 DOI: 10.1097/ms9.0000000000003217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/17/2025] [Indexed: 05/09/2025] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a complex endocrine disorder affecting 5%-10% of reproductive-aged women. Its diverse clinical spectrum includes reproductive, metabolic, and dermatological abnormalities, with cutaneous manifestations often serving as visible indicators of underlying hormonal and metabolic imbalances. Objective This review explores the pathophysiology, diagnosis, management, and psychosocial impact of the cutaneous manifestations of PCOS, providing an integrated understanding of their clinical significance. Methods A comprehensive analysis was conducted based on existing literature to elucidate the underlying mechanisms, diagnostic approaches, and treatment options for dermatological features associated with PCOS, including acne, hirsutism, acanthosis nigricans, seborrheic dermatitis, and androgenic alopecia. Results The pathophysiology of cutaneous manifestations in PCOS is driven by hyperandrogenism, insulin resistance, and local androgenic effects on the pilosebaceous unit. Acne and hirsutism are among the most common skin findings, followed by androgenic alopecia and acanthosis nigricans. Diagnostic strategies combine clinical evaluation with hormonal assays and imaging. Management requires a multidisciplinary approach encompassing hormonal therapies, lifestyle modifications, and targeted dermatological treatments. Additionally, these manifestations significantly impair psychosocial well-being, necessitating holistic care. Conclusion Cutaneous manifestations are a cosmetic concern and an essential diagnostic and therapeutic focus in PCOS. Addressing these features can enhance patient outcomes by mitigating physical symptoms and improving quality of life. Future research should emphasize personalized treatments and the psychosocial aspects of care to provide comprehensive management for women with PCOS.
Collapse
Affiliation(s)
- Muhammad Farhan
- Ajman University, College of Medicine, Ajman, United Arab Emirates
| | - Ariana Seyfi
- Ajman University, College of Medicine, Ajman, United Arab Emirates
| | - Afra Alnuaimi
- Dermatology Department, Sheikh Khalifa Medical Center, Abu Dhabi, United Arab Emirates
| | - Maya Alamour
- Ajman University, College of Medicine, Ajman, United Arab Emirates
| | - Sarah Alwarafi
- Ajman University, College of Medicine, Ajman, United Arab Emirates
| | - Haya Elastal
- Canadian Specialist Hospital, Dubai, United Arab Emirates
| | | | | | | | | | | | - Tirath Patel
- Trinity Medical Sciences University School of Medicine, St. Vincent, Saint Vincent and the Grenadines
| |
Collapse
|
29
|
Heydari T, Ramdass PV. Circadian rhythm disruption and polycystic ovary syndrome: a systematic review and meta-analysis. AJOG GLOBAL REPORTS 2025; 5:100479. [PMID: 40292350 PMCID: PMC12032313 DOI: 10.1016/j.xagr.2025.100479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Objective The aim of our systematic review and meta-analysis was to determine if circadian rhythm disruption (CRD) is associated with polycystic ovary syndrome (PCOS). Our objective was to pool the overall mean differences in biomarkers of CRD (including melatonin levels, morning and evening cortisol levels, and sleep efficiency) between PCOS patients and controls. We hypothesized that CRD will be more prominent in patients with PCOS. Data sources A systematic search of PubMed, Scopus, Embase, ClinicalTrials.Gov, and the Cochrane Database of Systematic Reviews was conducted from inception until 2024 using the following MeSH terms "circadian rhythm" OR "sleep disturbance" OR "melatonin" AND "polycystic ovary syndrome." Citation search supplemented the systematic database search. Study eligibility criteria Inclusion criteria were women with PCOS, original case-control, cross-sectional, and cohort studies that identify parameters of CRD (melatonin, cortisol, and sleep disturbance). Exclusion criteria were women with endocrine and metabolic co-morbidities, menopausal women, case reports, review studies, animal studies, abstracts, and conference presentations. There was no time restriction for year of publication. Study appraisal and synthesis methods Two investigators (T.H. and P.R.) assessed the quality of the studies included using the Newcastle-Ottawa Scale. Forest plots were created using the Open Meta Analyst software. Publication bias was assessed in Egger's and Begg's tests. Results A total of 16 studies were included in the systematic review and 12 studies were included in the meta-analysis (N=1,100 women [531 PCOS patients and 569 controls]). Pooled analysis showed that the mean difference in melatonin levels between PCOS patients and controls was 14.294 pg/mL, 95% CI [6.895, 21.693]. The overall mean difference in morning and evening cortisol between PCOS patients and controls was 1.103 pg/mL, 95% CI [-1.058, 3.265], and 3.574 pg/mL, 95% CI [1.741, 5.407], respectively. Pooled difference in mean sleep efficiency scores between PCOS patients and controls was -4.059, 95% CI [-6.752, -1.366]. Risk of bias assessment showed that NOS scores ranged from 7 to 9. Conclusions Our meta-analysis provides evidence that circadian rhythm disruption is positively associated with polycystic ovary syndrome. This is substantiated by differences in parameters indicative of circadian rhythm disruption, including melatonin levels, evening cortisol, and sleep efficiency.
Collapse
Affiliation(s)
- Tara Heydari
- Department of Public Health and Preventive Medicine (Heydari, and Ramdass), School of Medicine, St. George's University, St. George's, Grenada
| | - Prakash V.A.K. Ramdass
- Department of Public Health and Preventive Medicine (Heydari, and Ramdass), School of Medicine, St. George's University, St. George's, Grenada
| |
Collapse
|
30
|
Hakemi L, Norouzi G, Bakhshi A, Kazerouninejad E, Goldoozian M. Frequency of polycystic ovarian morphology and other ultrasonographic findings in Iranian female athletes. J Sci Med Sport 2025; 28:345-349. [PMID: 39799059 DOI: 10.1016/j.jsams.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVES Polycystic ovary morphology and ovarian cysts are frequent and usually asymptomatic in young female individuals. The present study is the first to examine ovarian sonographic findings in asymptomatic female athletes from Iran. This study aimed to evaluate the frequency of such sonographic findings in healthy female athletes on a screening basis. METHODS In this cross-sectional observational study, 455 females from 16 provinces of Iran were evaluated by trans-abdominal sonography during their routine pre-participation medical evaluation. RESULTS The mean age of participants was 21.8 years (SD 5.3). The mean age at the beginning of exercise was 14.20 years (SD = 4.25). Menarche occurred at 11-20 years with a mean of 13.5 years (SD 1.5). Judokas had the lowest (12.1 years), and football players had the highest menarche age (14.6 years; as compared to 12.8 years in the Iranian female population). In 87.5 % of participants, the sonographic findings were within normal limits. 4.8 % of all examinations revealed polycystic ovary morphology (PCOM), 5.7 % of participants had a simple cyst(s), and 20.8 % of cysts had diameters >49 mm. In those athletes who began regular performance-oriented training before menarche, PCOM was found in only 2.4 %. There was a significant positive linear association between BMI groups and PCOM (p = 0.003), but not for simple cysts. Frequency of PCOM (p < 0.001) as well as ovarian cysts (p = 0.012) were significantly different among studied sport types. There was a significant positive correlation between delayed menarche status and PCOM (p = 0.017). However, no such relationship was found for simple cysts. CONCLUSIONS Maintaining a healthy body mass index and beginning regular exercise before menarche may have a preventative effect for polycystic ovary morphology in female athletes.
Collapse
Affiliation(s)
- Laleh Hakemi
- Women Affairs, Sports Medicine Federation, Iran.
| | | | - Aida Bakhshi
- Faculty of Medicine, Mashhad University of Medical Sciences, Iran
| | | | - Maral Goldoozian
- Student of Biotechnology, Department of Biology, Islamic Azad University, Iran
| |
Collapse
|
31
|
Bui LM, Zaborek J, Eglash A, Cooney LG. Obesity but Not Polycystic Ovary Syndrome Associated with Decreased Breastfeeding Initiation Rates. Breastfeed Med 2025; 20:327-337. [PMID: 39932796 DOI: 10.1089/bfm.2024.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Objective: To determine whether women with polycystic ovary syndrome (PCOS) were less likely to initiate breastfeeding or have shorter breastfeeding duration. Materials and Methods: Cross-sectional analysis was performed of the Pregnancy Risk Assessment Monitoring System dataset, a national questionnaire from the Centers for Disease Control and Prevention of postpartum women, from 2016 to 2018. Logistic regression assessed the odds of any breastfeeding initiation. Cox proportional hazards assessed duration of any breastfeeding. Results: Sample included 2,382,290 women (6.1% PCOS). In univariable analysis, PCOS was associated with increased odds of any breastfeeding initiation (89.9% versus 87.9%; odds ratio [OR]: 1.23 [95% confidence interval: 1.02, 1.47]; p = 0.03). This outcome remained significant after controlling for body mass index (BMI; ORadj: 1.3 [1.1, 1.6]; p = 0.005) but not after controlling for education and prior live births (ORadj: 1.10 [0.89, 1.37]; p = 0.37). With increasing BMI, the odds of any breastfeeding initiation decreased, with the lowest odds seen in women with class III obesity (ORadj: 0.74 [0.60, 0.9]; p = 0.003). In a subanalysis of racial/ethnic groups, PCOS did not impact any breastfeeding initiation in White or Black non-Hispanic groups but increased odds of any breastfeeding initiation in Hispanic women (ORadj: 2.0 [1.1, 3.7]; p = 0.03). In multivariable models, there was no difference in the duration of any breastfeeding in women with PCOS compared with those without. Conclusions: Understanding predictors of breastfeeding success is paramount. In this national survey measuring any breastfeeding, PCOS did not decrease breastfeeding initiation or duration, despite confirming the association between overweight/obesity and decreased breastfeeding. However, because the data did not distinguish between exclusive breastfeeding and supplementation, we cannot rule out the possibility that PCOS affects breastfeeding exclusivity or necessitates supplementation. This limitation suggests that important trends could be obscured, and therefore, our findings should be interpreted with caution regarding breastfeeding exclusivity. Interventions aimed at increasing breastfeeding should target populations that would benefit the most; our data support that PCOS-specific targeting is not needed. Additional prospective studies are necessary to fully understand the association between different PCOS phenotypes and breastfeeding.
Collapse
Affiliation(s)
- Leeann M Bui
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA
| | - Jen Zaborek
- Department of Biostatistics, University of Wisconsin, Madison, Wisconsin, USA
| | - Anne Eglash
- Department of Family and Community Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Laura G Cooney
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
32
|
Shi W, Lin H, Di W, He C, Shen Y. Granulosa cell RNA-Seq insights into senescence and sphingolipid metabolism disorder in PCOS: aspirin as a potential therapeutic drug. Reprod Biol Endocrinol 2025; 23:61. [PMID: 40287692 PMCID: PMC12032776 DOI: 10.1186/s12958-025-01396-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a pivotal cause of anovulatory infertility and the pathogenesis remains elusive. Cellular senescence and sphingolipid metabolism disorder are closely intertwined, and both have been demonstrated present within the granulosa cells of PCOS, while research on the combined impact of senescence and sphingolipids on PCOS-related anovulation is scarce. METHODS Here, we leveraged four datasets of PCOS and executed differential gene expression analysis, engaged in WGCNA, and harnessed machine learning algorithms-including RF, SVM-RFE, and LASSO-to deeply explore the key genes that interact with senescence and sphingolipid metabolism in granulosa cells of PCOS. These key genes were subjected to further analysis to construct a diagnostic model, forecast immune cell infiltration, and identify potential agents. Additionally, within the testosterone-stimulated granulosa cells, we validated the expression of key genes, confirmed senescence and sphingolipids dysregulation, and evaluated the therapeutic efficacy of the candidate agent. RESULTS Our research pinpointed a set of genes (LYN, PLCG2, STAT5B, MMP9, and IL6R) that showed promise as biomarkers for PCOS-related anovulation and the diagnostic nomogram was developed. These biomarkers were linked to various immune cell types infiltration. In testosterone-stimulated granulosa cells, we observed increased expression of these biomarkers, accompanied by signs of senescence and changes in sphingolipids. Importantly, the potential agent aspirin displayed the ability to ameliorate these two processes. CONCLUSION This study highlighted the important value of genes associated with senescence and sphingolipids dysregulation in PCOS. Aspirin targeting senescence could be a promising therapeutic drug for addressing anovulation associated with PCOS.
Collapse
Affiliation(s)
- Weiwei Shi
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hao Lin
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Clinical Science and Research, Zhongda Hospital Affiliated to Southeast University, Nanjing, 21009, Jiangsu, China
| | - Wu Di
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Cong He
- Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing, 210013, Jiangsu, China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
33
|
Şener R, Alataş SE, Güler ÖT. Evaluation of serum calprotectin levels in patients with polycystic ovary syndrome. Sci Rep 2025; 15:14471. [PMID: 40281084 PMCID: PMC12032230 DOI: 10.1038/s41598-025-99445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age. PCOS is associated with many chronic complications such as endometrial thickening, endometrial cancer, breast cancer, infertility, dyslipidemia, and long-term cardiovascular disease. Therefore, many parameters and markers should be used in the evaluation of patients diagnosed with PCOS. Calprotectin has also been shown to be an important marker of destruction in many diseases that progress with inflammatory processes. The objective of our study was to examine the diagnostic efficacy of calprotectin in the context of polycystic ovary syndrome. In this study, 39 patients diagnosed with polycystic ovary syndrome and 41 healthy controls were analyzed. Serum calprotectin levels were compared between patients with different phenotypes of PCOS and a control group. Our study demonstrates significant differences in hormonal and inflammatory parameters between PCOS patients and healthy controls. PCOS patients exhibited significantly lower FSH levels and higher LH levels, resulting in a marked increase in the LH/FSH ratio. Additionally, DHEA-S and total testosterone levels were significantly elevated in the PCOS group, while SHBG levels were notably lower. The free androgen index (FAI) and serum calprotectin levels were also significantly higher in the PCOS group, highlighting the potential role of calprotectin in the pathophysiology of the disorder. Our study demonstrated that serum calprotectin levels are significantly higher in patients with PCOS compared to healthy controls. This finding underscores the potential role of calprotectin in the pathophysiology of PCOS and suggests that this marker could provide valuable insights into the inflammatory processes associated with the condition. However, further large-scale and long-term studies are necessary to better understand the impact of calprotectin on different PCOS phenotypes and its potential application in clinical practice.
Collapse
Affiliation(s)
- Rıfat Şener
- Department of Gynecology and Obstetrics, Gynecological Oncology Surgery, Konya City Hospital, Konya, Turkey.
| | - Süleyman Erkan Alataş
- Department of Gynecology and Obstetrics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Ömer Tolga Güler
- Department of Gynecology and Obstetrics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
34
|
Mohammed S, Sundaram V, Cockburn BN, Motilal S, Ottey S, Azziz R. The Association of Polycystic Ovary Syndrome-Like Clinical Features and Socioeconomic Status on Health-Related Quality of Life. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2025; 6:493-503. [PMID: 40538676 PMCID: PMC12177319 DOI: 10.1089/whr.2025.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 06/22/2025]
Abstract
Background Polycystic Ovary Syndrome (PCOS) affects 8%-13% of reproductive-age women globally, with comorbidities including obesity, insulin resistance, type 2 diabetes, and psychological disorders. Socioeconomic status (SES) significantly impacts health outcomes. Methods A community-based, pilot study was conducted in Trinidad among females aged 18-45 years, representing diverse ethnicities and SES. Participants underwent a standardized history and physical exam. Clinical hyperandrogenism (HIR) was assessed using the modified Ferriman-Gallwey scale (HIR ≥6), menstrual dysfunction (MD) as <9 cycles/year, depression via Beck's Inventory, overall health using SF-12 v1, and daytime somnolence with the Epworth Sleepiness Scale. Data analysis included descriptive statistics, analysis of variance, and multinomial logistic regression adjusting for confounders. Results Among 250 participants (mean age 31.6 ± 7.9 years), we classified 200 with clinical presentations, which included: no MD or HIR (56.7%), MD only (14.4%), HIR only (21.9%), and MD+HIR (7%). Age, income, and education were significantly correlated with clinical presentation. Older age reduced the risk of HIR (mean difference = 4.507, p = 0.004) and MD+HIR (mean difference = 9.063, p < 0.001). Income (OR = 0.37, 95% CI: 0.16-0.87, p = 0.022) reduced MD odds. Self-reported infertility was associated with MD (odds ratio [OR] = 0.27, 95% confidence interval [CI]: 0.11-0.65, p = 0.006). MD+HIR correlated with severe depression (OR = 5.96, 95% CI: 1.62-21.90, p = 0.007). Mental health scores (SF-12 MCS) were lower in women with MD+HIR (mean difference = -11.477, p = 0.005). Conclusion Seven percent of women in this sample showed probable PCOS based on clinical manifestations, with SES impacting quality of life, mental health, and sleep. Higher income reduced MD and MD+HIR risk, while infertility increased MD risk and severe depression was linked to MD+HIR.
Collapse
Affiliation(s)
- Stephanie Mohammed
- Dept. of Preclinical Sciences, Faculty of Medical Sciences, The University of the West Indies at St Augustine, Republic of Trinidad and Tobago
- Dept. of Physics, Faculty of Science and Technology, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Venkatesan Sundaram
- Dept. of Basic Veterinary Sciences, School of Veterinary Medicine, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Brian N. Cockburn
- Dept. of Life Sciences, Faculty of Science and Technology, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Shastri Motilal
- Dept. of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Sasha Ottey
- PCOS Challenge: The National Polycystic Ovary Syndrome, Atlanta, Georgia, USA
| | - Ricardo Azziz
- Dept. of Health Policy, Management, and Behavior, School of Public Health, University at Albany, SUNY, Rensselaer, New York, USA
- Depts. of Obstetrics & Gynecology, Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Dept. of Healthcare Organization & Policy, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
35
|
Ran B, Liu C, He Y, Ma L, Wang F. Bibliometric analysis of the research on anti-Müllerian hormone and polycystic ovary syndrome: current status, hotspots, and trends. FRONTIERS IN REPRODUCTIVE HEALTH 2025; 7:1519249. [PMID: 40342310 PMCID: PMC12058747 DOI: 10.3389/frph.2025.1519249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Background Polycystic Ovary Syndrome (PCOS) is a common endocrine and metabolic disorder affecting women of reproductive age. Over the past 30 years, significant efforts have been devoted to exploring its various pathogenic mechanisms, physiological and pathological characteristics, and biomarkers. Among these, Anti-Müllerian Hormone (AMH), as a biomarker for PCOS, is a significant biomarker for diagnosing, treating, and monitoring. However, the individual key information extracted from numerous studies is difficult to apply in clinical practice. Therefore, this article employs bibliometric analysis to summarize the current state of knowledge and offer future perspectives. Methods The Science Citation Index Expanded (SCI-E) within the Web of Science Core Collection database has been identified as the material source for obtaining articles related to AMH and PCOS. Software such as Origin, Microsoft Excel, Pajek, VOSviewer, and CiteSpace were used for bibliometric analysis and statistical assessment, evaluating countries, institutions, journals, references, and authors, as well as for constructing visual knowledge network maps. Results From 1994 to 2024, a total of 1,082 articles were included in the bibliometric analysis of research on AMH and PCOS. The number of publications in this field has consistently increased, with contributions from 70 countries, 1,363 institutions, and 5,144 researchers worldwide. Among them, the United States and China are the two countries with the highest number of publications. Zhejiang University, Monash University, and Peking University rank among the top three institutions exhibiting explosive citation bursts. The author with the highest publication volume is Didier Dewailly. The predictive keywords associated with these articles include "consensus," "morphology," "criteria," "prevalence," and "Müllerian hormone." Conclusions Through bibliometric analysis, this study has identified the primary research hotspots in the field of AMH and PCOS as follows: (1) Refining the diagnostic criteria for PCOS by using AMH as a biomarker; (2) Exploring the molecular role of AMH in the pathophysiological processes of various PCOS phenotypes and its potential as a therapeutic target; (3) Analyzing the impact of baseline AMH levels on female reproductive health and other biomarkers; (4) Investigating the signalling mechanisms of AMH in PCOS and its role in disease progression.
Collapse
Affiliation(s)
- Bingqing Ran
- School of Integrated Chinese and Western Clinical Medicine, Gansu University of Chinese Medicine, Research Experimental Center, Gansu Province, Lanzhou City, China
| | - Cai Liu
- Lanzhou University, School of Clinical Medicine, Gansu Province, Lanzhou City, China
| | - Yajun He
- Lanzhou University, School of Clinical Medicine, Gansu Province, Lanzhou City, China
| | - Lizhu Ma
- The Second Hospital, Lanzhou University, Department of Reproductive Medicine, Gansu Province, Lanzhou City, China
| | - Fang Wang
- The Second Hospital, Lanzhou University, Department of Reproductive Medicine, Gansu Province, Lanzhou City, China
| |
Collapse
|
36
|
Mei Y, Li W, Chen Z, Wang M. The association between serum growth differentiation factor 15 and insulin resistance in women diagnosed with polycystic ovary syndrome. Sci Rep 2025; 15:13824. [PMID: 40263510 PMCID: PMC12015211 DOI: 10.1038/s41598-025-98028-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is strongly associated with metabolic abnormalities, with 50-70% of patients exhibiting insulin resistance (IR), which significantly impacts the reproductive health of women in their reproductive years. Growth differentiation factor 15 (GDF15), a hormone responsive to nutritional stress, has been implicated in several diseases. This study sought to clarify the relationship between GDF15 levels and IR condition in PCOS patients. Based on the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), patients were categorized into an IR-PCOS group (n = 124) and a non-insulin-resistant group (non-IR-PCOS group, n = 109). Fasting blood samples were collected to measure GDF15 concentrations. To assess metabolic complications in relation to GDF15 levels, patients were also classified into high and normal GDF15 groups. Serum GDF15 levels were significantly higher in IR-PCOS patients (median 772.94 pg/ml) compared to non-IR-PCOS patients (median 575.80 pg/ml, P < 0.05). The high GDF15 group showed more severe metabolic and lipid abnormalities than the normal GDF15 group. Spearman correlation analysis revealed a correlation between increased GDF15 levels and impaired glucose metabolism. Logistic regression analysis identified GDF15, HDL-C, and prolactin as risk factors for IR in PCOS, and the fully adjusted regression coefficient for GDF15 levels and IR prevalence was 4.490 (95% CI 1.541 to 13.088). Restricted cubic spline analysis confirmed a positive association between GDF15 levels and IR within a specific range. The combined predictive probability of GDF15, prolactin, and HDL-C for IR was 0.763 (95% CI 0.701 to 0.826) according to ROC analysis. Elevated GDF15 levels may be associated with IR in PCOS patients, suggesting a potential role for GDF15 in the pathophysiology of IR in this condition.
Collapse
Affiliation(s)
- Yufeng Mei
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Wanzhen Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zhenni Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
37
|
Huang L, Guo Z, Jiang Z, Xu Y, Huang H. Resting metabolic rate in obesity. Postgrad Med J 2025; 101:396-410. [PMID: 39561990 DOI: 10.1093/postmj/qgae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/17/2024] [Indexed: 11/21/2024]
Abstract
The prevalence of obesity has continued to rise, and obesity and its attendant metabolic disorders are major global health threat factors. Among the current interventions for obesity, none have demonstrated sustained efficacy in achieving long-term outcomes. So, the identification of therapeutic targets is of paramount importance in the advancement and sustainability of obesity. Resting metabolic rate (RMR) constitutes 60%-75% of total energy expenditure and serves a crucial function in maintaining energy balance. Nevertheless, there exists considerable heterogeneity in RMR among individuals. Low RMR is associated with weight gain, elevating the susceptibility to obesity-related ailments. Hence, RMR will be the main focus of interest in the study of obesity treatment. In this review, we will elucidate the influence factors and mechanisms of action of RMR in obesity, with particular emphasis on the effects of obesity treatment on RMR and the alterations and influence factors of RMR in special types of populations with obesity.
Collapse
Affiliation(s)
- LingHong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, NO. 950 Donghai Street, Fengze District, Quanzhou 362000, Fujian, China
| | - ZhiFeng Guo
- Department of Respiratory Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, NO. 950 Donghai Street, Fengze District, Quanzhou 362000, Fujian, China
| | - ZhengRong Jiang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, NO. 950 Donghai Street, Fengze District, Quanzhou 362000, Fujian, China
| | - YaJing Xu
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, NO. 950 Donghai Street, Fengze District, Quanzhou 362000, Fujian, China
| | - HuiBin Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, NO. 950 Donghai Street, Fengze District, Quanzhou 362000, Fujian, China
| |
Collapse
|
38
|
Darand M, Ghorbani M, Ghadiri-Anari A, Arabi V, Hosseinzadeh M. The association between meat consumption and polycystic ovary syndrome in Iranian women: a case-control study. BMC Womens Health 2025; 25:189. [PMID: 40247277 PMCID: PMC12007253 DOI: 10.1186/s12905-025-03695-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND In light of the observed association between nutritional factors and polycystic ovary syndrome (PCOS) in recent decades, the present study was conducted to investigate the association between the consumption of various types of meat and PCOS in Iranian women. MATERIAL AND METHODS This frequency-matched case-control study included 108 women with newly diagnosed PCOS and 108 age and body-mass-index-matched women without PCOS, as a control group, who were referred to the Yazd Diabetes Clinic and Khatam Clinic between January 2018 and March 2019. The validated 178-item food frequency questionnaire was used to assess the usual dietary intake. Logistic regression analysis was used to estimate the association between meat consumption and PCOS. RESULTS The findings of this study showed, the individuals in the third tertile of red meat intake, had higher odds of PCOS in the crude model (Odds Ratio (OR) = 4.29; 95% Confidence Interval (CI), 2.13-8.64; P-value = 0.001) compared with those in the first tertile. These results remained significant after adjustments for energy intake, marital status, physical activity, education, pregnancy history and chronic disease history (OR = 3.87; 95% CI, 1.78-8.40; P-value = 0.001). Higher consumption of red meat increased the risk of PCOS by 3.87 times. Furthermore, higher consumption of processed meats increased the risk of PCOS by 2.15 times (OR = 2.15; 95% CI, 1.05-4.39; P-value and trend = 0.035). We did not find an association between other types of meat consumption and PCOS. CONCLUSION The results of the present study showed that a higher consumption of red and processed meat is associated with a higher risk of PCOS, whereas no significant correlation was found between the consumption of poultry, fish, and organ meat and PCOS. However, more studies are needed to support these findings in the future.
Collapse
Affiliation(s)
- Mina Darand
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moloud Ghorbani
- Department of Nutrition, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Akram Ghadiri-Anari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vahid Arabi
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Students' Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Hosseinzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Diabetes Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
39
|
Li M, He Z, Shi L, Lin M, Li M, Cheng Y, Liu H, Xue L, Said KS, Yusuf M, Galadanci HS, Nie L. Intelligent detection for Polycystic Ovary Syndrome (PCOS): Taxonomy, datasets and detection tools. Comput Struct Biotechnol J 2025; 27:1578-1599. [PMID: 40291542 PMCID: PMC12032871 DOI: 10.1016/j.csbj.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Recent research on Polycystic Ovary Syndrome (PCOS) detection increasingly employs intelligent algorithms to assist gynecologists in more accurate and efficient diagnoses. However, intelligent PCOS detection faces notable challenges: absence of standardized feature taxonomies, limited research on available datasets, and insufficient understanding of existing detection tools' capabilities. This paper addresses these gaps by introducing a novel analytical framework for PCOS diagnostic research and developing a comprehensive taxonomy comprising 108 features across 8 categories. Furthermore, we analyzed available datasets and assessed current intelligent detection tools. Our findings reveal that 12 publicly accessible datasets cover only 54% of the 108 features identified in our taxonomy. These datasets frequently lack multimodal integration, regular updates, and clear license information-constraints that potentially limit detection tool development. Additionally, our analysis of 42 detection tools identifies several limitations: high computational resource requirements, inadequate multimodal data processing, insufficient longitudinal analysis capabilities, and limited clinical validation. Based on these observations, we highlight critical challenges and future research directions for advancing intelligent PCOS detection tools.
Collapse
Affiliation(s)
- Meng Li
- Shenzhen Technology University, Shenzhen, Ghuangdong, China
| | - Zanxiang He
- Shenzhen Technology University, Shenzhen, Ghuangdong, China
| | - Liyun Shi
- Shenzhen People's Hospital, Shenzhen, Ghuangdong, China
| | - Mengyuan Lin
- Shenzhen People's Hospital, Shenzhen, Ghuangdong, China
| | - Minge Li
- Shenzhen People's Hospital, Shenzhen, Ghuangdong, China
| | - Yanjun Cheng
- Shenzhen People's Hospital, Shenzhen, Ghuangdong, China
| | - Hongwei Liu
- Shenzhen Technology University, Shenzhen, Ghuangdong, China
| | - Lei Xue
- Sun Yat-sen University, Guangzhou, Ghuangdong, China
| | | | | | | | - Liming Nie
- Shenzhen Technology University, Shenzhen, Ghuangdong, China
| |
Collapse
|
40
|
Hu R, Zhang L, Zhu J, Zhao S, Yin L, Hu J. Weight loss effects of non-pharmacological interventions in women with polycystic ovary syndrome: a systematic review and network meta-analysis. PeerJ 2025; 13:e19238. [PMID: 40256727 PMCID: PMC12009027 DOI: 10.7717/peerj.19238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 04/22/2025] Open
Abstract
Objective To compare the effectiveness of non-pharmacologic interventions in improving weight loss management in overweight patients with polycystic ovary syndrome (PCOS). Methods Five databases, PubMed, Embase, Cochrane, Web of Science and China Knowledge, were searched for this study. The Cochrane risk of bias tool was used to assess the risk of bias of eligible studies. The included randomized controlled trials were subjected to traditional meta-analysis (TMA) and network meta-analysis (NMA), and the cumulative number of surfaces under the ranking curve (SUCRA) was calculated for each intervention to derive the optimal intervention. Results The study ultimately included 29 articles involving 22 different interventions and 1,565 patients. The results of the NMA showed that the optimal intervention for the four outcome measures (body weight, body mass index (BMI), waist circumference (WC), waist-hip ratio (WHR)) was nutritional supplement + low-calorie diet, Taichi, continuous aerobic exercise and Taichi. Conclusion Current evidence suggests that nutritional supplements + hypocaloric diet; Taichi; continuous aerobic exercise have the greatest clinical advantage in weight loss and deserve to be promoted in the clinic. One of the best interventions for both outcome indicators, Taichi, suggests that it may be a common misconception that simply increasing the intensity of exercise is not the only way to lose weight and improve health.
Collapse
Affiliation(s)
- Rong Hu
- School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Lihong Zhang
- School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Zhu
- Medical College, Ankang University, Ankang, Shanxi, China
| | - Sihua Zhao
- The Second Ward of the Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lixue Yin
- Reproductive Center, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Junping Hu
- Reproductive Center, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
41
|
Ernest DK, Collier A, Chandrasekhar A, Xie L, Darraji S, Patel J, Almandoz JP, Messiah SE. Association of Polycystic Ovarian Syndrome Features and Metabolic Syndrome Among Reproductive-Aged Women in the United States. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2025; 6:431-441. [PMID: 40308359 PMCID: PMC12040555 DOI: 10.1089/whr.2024.0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 05/02/2025]
Abstract
Background Polycystic ovarian syndrome (PCOS) is associated with the metabolic health of racially and ethnically diverse women globally, but limited research exists on the association of PCOS and metabolic syndrome (MetS) among women in the United States. Objective To examine the association of PCOS features and MetS in a racially/ethnically diverse population of reproductive-aged women in the United States. Methods Cross-sectional data from 2,172 women (12-49 years) from the 2011-2016 National Health and Nutrition Examination Survey were analyzed. Univariate logistic regression models determined unadjusted associations of MetS and its components (elevated central obesity, glucose, blood pressure and triglyceride, and low high-density lipoprotein cholesterol) with PCOS features (log-transformed total testosterone (LTT), sex-hormone binding globulin (LSHBG), amenorrhea, and oral contraceptive pills (OCP) use). Multivariable logistic models examined age-adjusted associations stratified by race and ethnicity. Results The analytical sample (mean age = 30.3 years, 59% non-Hispanic White, 12.4% non-Hispanic Black, 18.7% Hispanic/Latina, 6.2% non-Hispanic Asian, 3.7% Other/multi-race) had a MetS prevalence of 14.5%. Overall, MetS was associated with age, body mass index, race/ethnicity, LTT and LSHBG concentrations, amenorrhea, and OCP use (p < 0.01 for all), and many of the PCOS features were protective against individual MetS components. Most race/ethnicities showed significantly lower odds of MetS with an increase in LSHBG, with varying impacts on individual MetS features. Conclusions Findings suggest significant associations between PCOS features and MetS among a racially and ethnically diverse population of reproductive-aged women in the United States. More robust and longitudinal studies are needed to further understand the underlying mechanism linking PCOS and MetS.
Collapse
Affiliation(s)
- Deepali K. Ernest
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Asha Collier
- School of Human Ecology, University of Texas at Austin, Austin, Texas, USA
| | - Aparajita Chandrasekhar
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Luyu Xie
- Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Jenil Patel
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
- School of Public Health, UTHealth at Houston, Dallas, Texas, USA
| | - Jaime P. Almandoz
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sarah E. Messiah
- Peter O’Donnell Jr School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
42
|
Sumer F, Gurlek B, Yildiz E, Uzun F, Colak S. Alterations in tear function and meibomian gland dysfunction across various phenotypes of polycystic ovary syndrome. Sci Rep 2025; 15:12680. [PMID: 40221508 PMCID: PMC11993714 DOI: 10.1038/s41598-025-94890-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
The aim of this study was to investigate the relationship between tear function changes and meibomian gland dysfunction in various phenotypes of polycystic ovary syndrome (PCOS). In this prospective case-control study, 4 separate case groups were formed for 4 different phenotypes of PCOS, which are variants of clinical presentation. A total of 160 women were included in the study, 32 women for each group and 32 healthy women as control group. Ocular surface disease index (OSDI), Oxford score for corneal and conjunctival involvement, Schirmer 1 and 2 tests, noninvasive tear breakup time (NIBUT), lower lid meibomian drop-out grades by non-contact meibography, and meibomian gland disease (MGD) distortion and shortening scores were compared between all groups. When the PCOS group was compared with the control group, phenotype A showed the most significant differences in NIBUT (p = 0.003), MGD (p = 0.002) and OSDI (p = 0.004). Although the mean Schirmer 1 and Schirmer 2 tests value were lower in the PCOS group compared to the control group, no significant difference was detected in any phenotype (p = 0.911, p = 0.117). A statistically significant weak negative correlation was observed between NIBUT and OSDI values (r = - 0.206, p = 0.027). There was a statistically significant positive and very weak correlation between OSDI and BMI (r = 0.079, p = 0.041). Tear film instability attributed to MGD was found to be significantly different in all PCOS patients compared to healthy subjects regardless of phenotype, and this difference was statistically higher in phenotype A.
Collapse
Affiliation(s)
- Fatma Sumer
- Department of Ophthalmology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100, Rize, Turkey.
| | - Beril Gurlek
- Department of Obsetrics and Gynaecology, Recep Tayyip Erdogan Faculty of Medicine, 53100, Rize, Turkey
| | - Elif Yildiz
- Department of Obsetrics and Gynaecology, İstanbul Gaziosmanpasa Training and Research Hospital, Istanbul, Turkey
| | - Feyzahan Uzun
- Department of Ophthalmology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100, Rize, Turkey
| | - Sabri Colak
- Department of Obsetrics and Gynaecology, Recep Tayyip Erdogan Faculty of Medicine, 53100, Rize, Turkey
| |
Collapse
|
43
|
Najdgholami Z, Sedgi FM, Ghalishourani SS, Feyzpour M, Rahimlou M. Flaxseed intervention and reproductive endocrine profiles in patients with polycystic ovary syndrome: an open-labeled randomized controlled clinical trial. Front Endocrinol (Lausanne) 2025; 16:1531762. [PMID: 40260281 PMCID: PMC12010250 DOI: 10.3389/fendo.2025.1531762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/13/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Polycystic Ovary Syndrome (PCOS), affecting 6-15% of women of reproductive age, is characterized by hormonal imbalances and metabolic disturbances. Flaxseed, rich in omega-3 fatty acids and phytoestrogens, may offer a natural approach to improve reproductive hormone profiles in PCOS patients. This study is aimed at evaluating the effects of flaxseed supplementation on reproductive endocrine profiles in women with PCOS. Methods In this open-labeled, randomized controlled trial, 70 women with PCOS were randomly assigned to either the intervention group (n=35), receiving 30 grams of milled flaxseed daily along with lifestyle modifications, or the control group (n=35), receiving only lifestyle modifications, for 12 weeks. The primary outcome was the change in follicle-stimulating hormone (FSH) levels. Secondary outcomes included changes in serum concentrations of luteinizing hormone (LH), the LH/FSH ratio, anti-Müllerian hormone (AMH), estradiol, dehydroepiandrosterone sulfate (DHEAS), and androstenedione. Results After 12 weeks, the flaxseed group showed a significant increase in FSH levels compared to the control group. FSH levels increased from 9.72 ± 11.95 µU/mL at baseline to 10.59 ± 12.14 µU/mL after the intervention (p = 0.027). The mean treatment effect was 0.87 (95% CI: 0.086 to 1.75). The LH/FSH ratio also significantly decreased in the flaxseed group (mean treatment effect: -0.341, 95% CI: -0.63 to -0.08, p = 0.031). No significant changes were observed in AMH, LH, estradiol, DHEAS, or androstenedione levels. Conclusion Flaxseed supplementation may benefit women with PCOS by increasing FSH levels and decreasing the LH/FSH ratio, suggesting its potential as a complementary intervention for managing hormonal disturbances in PCOS. Further studies are needed to confirm these findings and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Najdgholami
- Department of Midwifery, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fatemeh Maleki Sedgi
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Marzieh Feyzpour
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
- Metabolic Diseases Research Center, Health and Metabolic Research Institute, Zanjan University of Medical Science, Zanjan, Iran
| |
Collapse
|
44
|
Weiss NS, Kostova EB, Mol BWJ, van Wely M. Gonadotropins for ovulation induction in women with polycystic ovary syndrome. Cochrane Database Syst Rev 2025; 4:CD010290. [PMID: 40193219 PMCID: PMC11975188 DOI: 10.1002/14651858.cd010290.pub4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
RATIONALE Ovulation induction with follicle-stimulating hormone (FSH) is a second-line treatment in women with polycystic ovary syndrome (PCOS) who do not ovulate or conceive on clomiphene citrate or letrozole, though induction protocols and types of gonadotropins used vary greatly. OBJECTIVES To compare the effectiveness and safety of gonadotropins as a second-line treatment for ovulation induction in women with PCOS who do not ovulate or conceive after clomiphene citrate or letrozole. SEARCH METHODS In March 2024, we searched the Cochrane Gynaecology and Fertility Group Specialised Register of Controlled Trials, CENTRAL, MEDLINE, Embase and PsycINFO. We checked references of all relevant studies. We had no language or date restrictions. ELIGIBILITY CRITERIA All randomised controlled trials (RCTs) reporting data on clinical outcomes in women with PCOS who did not ovulate or conceive on clomiphene citrate or letrozole, and were undergoing ovulation induction with urinary-derived gonadotropins, including urofollitropin in purified FSH (uFSH) or highly purified FSH (HP-FSH) form, human menopausal gonadotropin (HMG) and highly purified human menopausal gonadotropin (HP-HMG), or recombinant FSH (rFSH) were eligible. We included trials reporting on ovulation induction followed by intercourse or intrauterine insemination. We excluded studies that described co-treatment with clomiphene citrate, metformin, luteinising hormone, or letrozole. OUTCOMES We implemented the core outcome set for infertility. Our critical outcomes were live birth rate and multiple pregnancy rate per woman. Important outcomes were clinical pregnancy, pregnancy loss, incidence of ovarian hyperstimulation syndrome (OHSS) per woman, total gonadotropin dose, total duration of stimulation per woman, gestational age at birth, birthweight, neonatal mortality, and major congenital anomaly. RISK OF BIAS We used the Cochrane RoB 1 tool to assess bias in the included studies. SYNTHESIS METHODS Where meta-analysis was possible, we combined data using a fixed-effect model to calculate the risk ratio (RR) or mean difference. We summarised the overall certainty of evidence for the main outcomes using GRADE criteria. INCLUDED STUDIES We included 15 studies with 2348 women. Ten trials compared rFSH with urinary-derived gonadotropins (one compared rFSH with human menopausal gonadotropin (HMG), and nine compared rFSH with urinary FSH). Three trials compared HMG with purified FSH (uFSH). One trial compared HP-FSH with purified FSH (uFSH) and one trial compared gonadotropins with continued clomiphene citrate. SYNTHESIS OF RESULTS Recombinant FSH (rFSH) versus urinary-derived gonadotropins There may be little or no difference in the birth rate between rFSH and urinary-derived gonadotropins (RR 1.21, 95% confidence interval (CI) 0.83 to 1.78; 5 RCTs, 505 participants; low-certainty evidence). This suggests that if the observed average live birth per woman who used urinary-derived gonadotropins is 16%, the chance of live birth with rFSH is between 13% and 28%. There may be little or no difference between groups in multiple pregnancy (RR 0.86, 95% CI 0.46 to 1.61; 8 RCTs, 1368 participants; low-certainty evidence), clinical pregnancy rate (RR 1.05, 95% CI 0.88 to 1.27; 8 RCTs, 1330 participants; low-certainty evidence), or miscarriage rate (RR 1.20, 95% CI 0.71 to 2.04; 7 RCTs, 970 participants; low-certainty evidence). We are uncertain whether rFSH reduces ectopic pregnancy (RR 2.81, 95% CI 0.12 to 67.90; 1 RCT, 151 participants; very-low certainty evidence) or the incidence of OHSS (RR 1.48, 95% CI 0.82 to 2.65; 10 RCTs, 1565 participants; very low-certainty evidence) when compared to urinary-derived gonadotropins. Human menopausal gonadotropin (HMG) versus purified urinary FSH (uFSH) When compared to uFSH, we are uncertain whether HMG improves live birth rate (RR 1.44, 95% CI 0.55 to 3.76; 2 RCTs, 79 participants), or reduces multiple pregnancy (RR 6.56, 95% CI 0.28 to 152.45; 3 RCTs, 102 participants). We are also uncertain whether HMG improves clinical pregnancy rate (RR 1.31, 95% CI 0.66 to 2.59; 3 RCTs, 102 participants), reduces miscarriage rate (RR 0.33, 95% CI 0.06 to 1.97; 2 RCTs, 98 participants), or reduces the incidence of OHSS (RR 7.07, 95% CI 0.42 to 117.81; 2 RCTs, 53 participants) when compared to uFSH. No trials reported on ectopic pregnancy. The certainty of the evidence was very low for all outcomes. Gonadotropins versus continued clomiphene citrate Gonadotropins (FSH) probably result in more live births than continued clomiphene citrate (RR 1.24, 95% CI 1.05 to 1.46; 1 RCT, 661 participants; moderate-certainty evidence). This suggests that for a woman with a live birth rate of 41% with continued clomiphene citrate, the live birth rate with gonadotropins was between 43% and 60%. There may be little or no difference in multiple pregnancy between treatments (RR 0.89, 95% CI 0.33 to 2.44; 1 RCT, 661 participants; low-certainty evidence). Gonadotropins probably result in more clinical pregnancies than continued clomiphene citrate (RR 1.31, 95% CI 1.13 to 1.52; 1 RCT, 661 participants; moderate-certainty evidence), and may result in more miscarriages (RR 2.23, 95% CI 1.11 to 4.47; 1 RCT, 661 participants; low-certainty evidence). We are uncertain if there is a difference in ectopic pregnancy between the groups (RR 0.51, 95% CI 0.09 to 2.77; 1 RCT, 661 participants; very low-certainty evidence). None of the women developed OHSS. The main limitations were imprecision, inconsistency, and indirectness. AUTHORS' CONCLUSIONS There may be little or no difference in live birth, multiple pregnancy, clinical pregnancy, or miscarriage rates between rFSH and uFSH in women with PCOS. For HMG versus uFSH, we are uncertain whether one or the other improves or lowers rates of live birth, multiple pregnancy, clinical pregnancy, or miscarriage. We are uncertain whether any of the interventions reduce ectopic pregnancy or the incidence of OHSS. In women with clomiphene citrate failure, gonadotropins (FSH) probably result in more live births and clinical pregnancies than continued clomiphene citrate without increasing multiple pregnancies. Gonadotropins may increase the miscarriage rate per woman. We are uncertain if gonadotropins reduce ectopic pregnancy. None of the women developed OHSS. FUNDING This Cochrane review had no dedicated funding. REGISTRATION Protocol (2012) https://doi.org/10.1002/14651858.CD010290 Review (2015) https://doi.org/10.1002/14651858.CD010290.pub2/full Update (2019) https://doi.org/10.1002/14651858.CD010290.pub3.
Collapse
Affiliation(s)
- Nienke S Weiss
- Gynaecology and Obstetrics, Amstelland Hospital, Amstelveen, Netherlands
| | - Elena B Kostova
- Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ben Willem J Mol
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Madelon van Wely
- Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
45
|
Zhu S, Huang Z, Chen X, Jiang W, Zhou Y, Zheng B, Sun Y. Construction and evaluation of machine learning-based prediction model for live birth following fresh embryo transfer in IVF/ICSI patients with polycystic ovary syndrome. J Ovarian Res 2025; 18:70. [PMID: 40186314 PMCID: PMC11969817 DOI: 10.1186/s13048-025-01654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE To investigate the determinants affecting live birth outcomes in fresh embryo transfer among polycystic ovary syndrome (PCOS) patients using various machine learning (ML) algorithms and to construct predictive models, offering novel insights for enhancing live birth rates in this specific group. METHODS A sum of 1,062 fresh embryo transfer cycles involving PCOS patients were analyzed, with 466 resulting in live births. The dataset was split randomly into training and testing subsets at a 7:3 ratio. Least absolute shrinkage and selection operator and recursive feature elimination methods were utilized for feature selection within the training data. A grid search strategy identified the optimal parameters for seven ML models: decision tree (DT), K-nearest neighbors (KNN), light gradient boosting machine (LightGBM), naive Bayes model(NBM), random forest (RF), support vector machine (SVM) and extreme gradient boosting (XGBoost). The evaluation of model effectiveness incorporated diverse metrics, encompassing area under the curve (AUC), accuracy, positive predictive value, negative predictive value, F1 score, and Brier score. Calibration curves and decision curve analysis were employed to ascertain the optimal model. Furthermore, Shapley additive explanations were applied to elucidate the importance of predictor variables in the top-performing model. RESULTS The AUC values of DT, KNN, LightGBM, NBM, RF, SVM and XGBoost models in the training set were 0.813, 1.000, 0.724, 0.791, 1.000, 0.819 and 0.853, respectively. Corresponding values in the testing set were 0.773, 0.719, 0.705, 0.764, 0.794, 0.806 and 0.822. XGBoost emerged as the most effective ML model. SHAP analysis revealed that variables encompassing embryo transfer count, embryo type, maternal age, infertility duration, body mass index, serum testosterone (T) levels, and progesterone (P) levels on the day of human chorionic gonadotropin administration were pivotal predictors of live birth outcomes in individuals with PCOS receiving fresh embryo transfer. CONCLUSION This study developed a live birth prediction model tailored for PCOS fresh embryo transfer cycles, leveraging ML algorithms to compare the efficacy of multiple models. The XGBoost model demonstrated superior predictive capacity, enabling prompt and precise identification of critical risk factors influencing live birth outcomes in PCOS patients. These findings offer actionable insights for clinical intervention, guiding strategies to improve pregnancy outcomes in this population. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Suqin Zhu
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China
- Fujian Maternal-Fetal Clinical Medicine Research Center, Fuzhou, 350001, China
| | - Zhiqing Huang
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China
- Fujian Key Laboratory of Prenatal Diagnosis and Birth Defect, Fuzhou, 350001, China
| | - Xiaojing Chen
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Wenwen Jiang
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Yuan Zhou
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Beihong Zheng
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China.
| | - Yan Sun
- Center of Reproductive Medicine, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou City, 350001, Fujian Province, China.
- Fujian Key Laboratory of Prenatal Diagnosis and Birth Defect, Fuzhou, 350001, China.
| |
Collapse
|
46
|
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous familial disorder affecting up to one in five women. The aetiology remains unclear, but available evidence suggests it is a polygenic disorder with epigenetic, developmental, and environmental components. The diagnostic criteria for PCOS are based on reproductive features, and the syndrome is categorized into several phenotypes that can vary by race and ethnicity. Insulin resistance and metabolic dysfunction have a crucial role in the pathogenesis of the syndrome and contribute to many adverse metabolic outcomes that place a substantial burden on the health of women with PCOS across their lifespan. Metabolic abnormalities like those identified in women with PCOS are also present in their female and male first-degree relatives. Overall, more emphasis is required on defining PCOS as a metabolic disorder in addition to a reproductive one. This approach could affect the management and future treatment options for the syndrome. The rationale of the current review is to identify and analyse existing evidence for PCOS as a metabolic, as well as a reproductive, disease.
Collapse
Affiliation(s)
- Nafiye Helvaci
- Division of Endocrinology and Metabolism, Ankara Ataturk Sanatoryum Training and Research Hospital, Ankara, Turkey
| | - Bulent Okan Yildiz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hacettepe University School of Medicine, Ankara, Turkey.
| |
Collapse
|
47
|
Tan Z, Wu T, Wang M, Chen L, Li Y, Zhang M, Zhang Y, Sun L. Downregulation of FASN in granulosa cells and its impact on ovulatory dysfunction in PCOS. J Ovarian Res 2025; 18:67. [PMID: 40170064 PMCID: PMC11959749 DOI: 10.1186/s13048-025-01645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/12/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complicated endocrinological and anovulatory disorder in women. Mice exposed to dihydrotestosterone (DHT) exhibit a PCOS-like phenotype characterized by abnormal steroid hormone production and ovulation dysfunction. The present investigation aims to identify overlapping genes expressed in PCOS patients and a PCOS mouse model induced by DHT and to examine the function of key genes fatty acid synthase (FASN) in hormone production and ovulation dysfunction. RESULTS We examined 5 datasets of high-throughput mRNA transcription from the Gene Expression Omnibus (GEO) database, including 4 datasets from individuals with PCOS and 1 dataset from a DHT-induced mouse model. GO and KEGG enrichment analyses revealed these differentially expressed genes (DEGs) are primarily involved in ovarian steroidogenesis and fatty acid metabolism. The PPI network identified 12 hub genes. qRT-PCR verification in human granulosa cells showed differential expression of FASN, SCARB1, FABP5, RIMS2, and RAPGEF4 in PCOS patients (p < 0.05). FASN was downregulated in the granulosa cells (GCs) of PCOS patients (p < 0.05). FASN depletion reduced KGN cell proliferation (p < 0.001), decreased progesterone secretion (p < 0.05), and increased estradiol secretion (p < 0.05). Downregulation of FASN inhibited ovulation by suppressing ERK1/2 phosphorylation and the expression of C/EBPα and C/EBPβ. Lentivirus-mediated FASN downregulation in rat ovaries for one and four weeks impaired the super ovulatory response, reducing oocyte retrieval, estrous cycle, secretion of estrogen and progesterone, and luteinization. CONCLUSIONS Our results provide new insights into PCOS pathogenesis and suggest that FASN could be a promising target for treating abnormal steroid hormone production and impaired ovulation in PCOS.
Collapse
Affiliation(s)
- Zhaoping Tan
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
- Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Tiancheng Wu
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
- Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Mei Wang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
- Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Liang Chen
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
- Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Yating Li
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
- Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Ming Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yuanzhen Zhang
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, Hubei, People's Republic of China.
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
| | - Lili Sun
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
48
|
Del Río JP, Tsompanidis A, Gaspar PA, Maturana-Hurtado A, Rojas-Costa GM, Dagnino-Subiabre A, Olea A, Maliqueo M, Echiburú B, de Guevara AL, Montiel JF, Baron-Cohen S, Crisosto N. Women with polycystic ovary syndrome (PCOS): Likelihood of cooccurring neuropsychiatric conditions and the dual hit hypothesis. Front Neuroendocrinol 2025; 77:101188. [PMID: 40120958 DOI: 10.1016/j.yfrne.2025.101188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Polycystic Ovary Syndrome (PCOS) is the most common endocrine-metabolic disorder in women of reproductive age. Hyperandrogenism has been proposed as its main pathophysiological feature. PCOS is associated with co-occurring conditions, including psychiatric disorders such as anxiety, depression, and neurodevelopmental conditions such as autism. Exposure to hyperandrogenism during prenatal life and adolescence may explain this association. PCOS women exhibit hyperandrogenism during pregnancy, and up to 70% of their daughters will present a similar phenotype from puberty onwards. The 'dual hit hypothesis' proposes that stressors during prenatal life and adolescence can synergistically lead to co-occurring conditions in adulthood. PCOS has been recently proposed as an independent likelihood factor for the development of neuropsychiatric conditions. However, the specific mechanisms require further research to develop effective interventions. This review discusses how hyperandrogenism can affect neurodevelopment during two key periods of brain development, which may explain the long-term impact of PCOS on mental health.
Collapse
Affiliation(s)
- Juan Pablo Del Río
- Department of Child and Adolescent Psychiatry and Mental Health, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Imhay. Santiago, Chile
| | - Alexandros Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, CB2 8AH, UK
| | - Pablo A Gaspar
- Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Imhay. Santiago, Chile; Psychiatry and Mental Health Department, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Alejandro Maturana-Hurtado
- Department of Child and Adolescent Psychiatry and Mental Health, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Gonzalo M Rojas-Costa
- School of Medicine, Finis Terrae University, 750115, Santiago, Chile; Biomedical Imaging Unit and Artificial Intelligence FISABIO-CIPF, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region, Valencia, Spain
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, CIESAL, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile; Millennium Institute for Depression and Personality Research (MIDAP), Santiago, Chile
| | - Arabia Olea
- Laboratory of Stress Neurobiology, CIESAL, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Bárbara Echiburú
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Amanda Ladrón de Guevara
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Juan F Montiel
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago 8370191, Chile
| | - Simon Baron-Cohen
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK; Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, CB2 8AH, UK
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Universidad del Desarrollo, Santiago 7610658, Chile.
| |
Collapse
|
49
|
Özgürlük İ, Kavak RP, Turhan B, Kaymak SD, Şeker R, Yeğin GF, Doğan İS. The relationship between spexin and liver steatosis in polycystic ovary syndrome: a novel analysis using ultrasound-guided attenuation parameter. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2025; 71:e20241313. [PMID: 40172394 PMCID: PMC11964304 DOI: 10.1590/1806-9282.20241313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 04/04/2025]
Abstract
OBJECTIVE This study aims to evaluate the relationship between spexin levels and nonalcoholic fatty liver disease in patients with polycystic ovary syndrome. METHODS The study included 90 participants, comprising 44 patients diagnosed with polycystic ovary syndrome and 46 age- and body mass index-matched controls. Participants' spexin, glucose, urea, alanine aminotransferase, aspartate aminotransferase, total cholesterol, low-density lipoprotein, high-density lipoprotein, triglycerides, progesterone, follicle-stimulating hormone, luteinizing hormone, estradiol, total testosterone, free testosterone, and dehydroepiandrosterone sulfate levels were measured. The liver steatosis grading was done using the ultrasound-guided attenuation parameter from General Electric Healthcare. The parameters were evaluated between the groups. RESULTS Spexin levels in polycystic ovary syndrome patients were significantly lower compared to the control group (p<0.001). Glucose, triglycerides, luteinizing hormone, total testosterone, free testosterone, and dehydroepiandrosterone sulfate levels were found to be significantly higher in polycystic ovary syndrome patients compared to the control group (p<0.001). High-density lipoprotein levels in polycystic ovary syndrome patients were significantly lower compared to the control group (p<0.001). The prevalence of liver steatosis was notably higher among the polycystic ovary syndrome patients compared to the control group (p<0.001). In patients with polycystic ovary syndrome, spexin levels demonstrated significant negative correlations with body mass index, glucose, triglycerides, luteinizing hormone, total testosterone, free testosterone, and dehydroepiandrosterone sulfate (all p<0.001). A strong negative correlation was observed between spexin levels and liver steatosis grading (p<0.001). CONCLUSION Spexin is a coordinator in the metabolic relationship between polycystic ovary syndrome and liver health, suggesting its utility as a biomarker for detecting liver steatosis and related metabolic disturbances in this population.
Collapse
Affiliation(s)
- İzzet Özgürlük
- Ministry of Health, Etlik City Hospital, Department of Clinic of Obstetrics and Gynecology – Ankara, Turkey
| | - Rasime Pelin Kavak
- Ministry of Health, Etlik City Hospital, Department of Radiology – Ankara, Turkey
| | - Berna Turhan
- Ministry of Health, Etlik City Hospital, Department of Radiology – Ankara, Turkey
| | - Sümeyya Duran Kaymak
- Ministry of Health, Etlik City Hospital, Department of Radiology – Ankara, Turkey
| | - Rabia Şeker
- Ministry of Health, Etlik City Hospital, Department of Medical Biochemistry – Ankara, Turkey
| | - Gülin Feykan Yeğin
- Ministry of Health, Etlik City Hospital, Department of Clinic of Obstetrics and Gynecology – Ankara, Turkey
| | - İhsaniye Süer Doğan
- Ministry of Health, Etlik City Hospital, Department of Radiology – Ankara, Turkey
| |
Collapse
|
50
|
Almotairi R, Mir R, Almasoudi KS, Husain E, Mtiraoui N. Evaluating the Impact of rs4025935, rs71748309, rs699947, and rs4646994 Genetic Determinants on Polycystic Ovary Syndrome Predisposition-A Case-Control Study. Life (Basel) 2025; 15:558. [PMID: 40283113 PMCID: PMC12028387 DOI: 10.3390/life15040558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Background: As a complicated endocrine condition, polycystic ovarian syndrome affects around 20% of women who are of reproductive age. It is linked to an increased risk of endometrial cancer, cardiovascular diseases, mental illnesses, non-alcoholic fatty liver disease, metabolic syndrome, and Type 2 diabetes. Despite numerous genetic studies identifying several susceptibility loci, these only account for approximately 10% of the hereditary factors contributing to PCOS, leaving its etiology largely unknown. While genome-wide association studies (GWAS) have been conducted on various populations to identify SNPs linked to PCOS risk, no such study has been reported in Tabuk. Thus, this study aims to investigate the association of a glutathione S-transferase M1 (GSTM1) deletion, VEGF gene (I/D) insertion/deletion, and VEGF-2578 gene polymorphism with polycystic ovarian syndrome. Methodology: In this research study (case-control), we utilized the ARMS-PCR to determine and analyze the polymorphic variants of VEGF-2578 C/A (rs699947). We employed multiplex PCR for the GSTM1 deletion and MS-PCR (mutation specific PCR) for the vascular endothelial growth factor gene insertion/deletion. Results: The findings indicated statistically significant differences in various biochemical and endocrine serum biomarkers, including lipid profiles (cholesterol, HDL, and LDL), Type 2 diabetes markers (HOMA-IR (Homeostatic Model Assessment for Insulin Resistance), free insulin fasting glucose), and hormone levels (testosterone, LH, progesterone and FSH) in PCOS patients. Specifically, regarding the GSTT1 genotype, individuals with the GSTT1-null genotype had an odds ratio (OR) of 4.16 and a relative risk (RR) of 2.14 compared to those with the GSTT1 genotype, with statistically significant differences (p = 0.0001). However, for the GSTM1 genotype, there was a statistically significant difference (p = 0.0002) in the OR and RR for the GSTM1-null genotype, which were 2.66 and 1.64, respectively. Protective effects were observed for individuals with either GSTT1 (+) GSTM1 (-) or GSTT1 (-) GSTM1 (+) genotypes, as well as for those with both null genotypes, yielding an OR of 0.41 and p < 0.003. The VEGF rs699947 C>A gene variation showed a statistically significant association between PCOS patients and controls (p < 0.020), with the A allele frequency higher among PCOS patients (0.42 vs. 0.30). Similarly, the VEGF rs4646994 I>D gene variation exhibited a statistically significant difference (p < 0.0034), with the D allele being more frequent in PCOS patients (0.52 vs. 0.35). The VEGF-A allele was strongly linked to PCOS susceptibility in the allelic model, exhibiting an OR of 1.62, RR of 1.27, and p < 0.007, while in the allelic comparison, the OR was 1.71, the RR was 1.32, and p < 0.004. Conclusions: This study concluded that null genotypes at rs4025935 and rs71748309, an insertion deletion at rs4646994, and the A allele of rs699947 were significantly associated with PCOS predisposition in our population and these could serve as potential loci for PCOS predisposition. To the best of our knowledge, it is the first study to highlight the association between these genetic variations and the predisposition of PCOS in our populations. Large-scale case-control studies in the future are required to confirm these results.
Collapse
Affiliation(s)
- Reema Almotairi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia; (K.S.A.); (E.H.)
| | - Rashid Mir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia; (K.S.A.); (E.H.)
| | - Kholoud S. Almasoudi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia; (K.S.A.); (E.H.)
| | - Eram Husain
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia; (K.S.A.); (E.H.)
| | - Nabil Mtiraoui
- Laboratory of Human Genome and Multifactorial Diseases, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia;
| |
Collapse
|