1
|
Eltokhy AK, El-Shaer RAA, El-Deeb OS, Farghal EE, Ibrahim RR, Elesawy R, Awad MM, Ismail R, Motawea SM, Shatat D, Hafez YM, El Hanafy HA, Atef MM. Synergistic effects of AgNPs and zileuton on PCOS via ferroptosis and inflammation mitigation. Redox Rep 2025; 30:2445398. [PMID: 39723580 DOI: 10.1080/13510002.2024.2445398] [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: 12/28/2024] Open
Abstract
BACKGROUND The most prevalent endocrine disorder affecting women is PCOS. Programmed death of ovarian cells has yet to be elucidated. Ferroptosis is a kind of iron-dependent necrosis featured by significantly Fe+2-dependent lipid peroxidation. The ongoing study aimed to reinforce fertility by combining therapy with AgNPs and (Zileuton) in PCOS rats' model. METHODS The study included 75 adult female rats divided into 5 groups; control, PCOS, PCOS treated with AgNPs, PCOS treated with Zileuton, and PCOS group treated with AgNPs and Zileuton. The study investigated the anti-ferroptotic, anti-inflammatory, antioxidant, antiapoptotic, histopathological and immunohistochemical examinations of COX-2 and VEGF. RESULTS The combination of AgNPs and Zileuton showed significant reduction of inflammatory mediators (IL-6, TNF-α, NFk-B) compared with diseased group (P-value < 0.05), regression of ferroptosis marks (Panx1 and TLR4 expression, Fe+2 levels) compared with diseased group (P-value < 0.05), depression of apoptotic marker caspase 3 level compared with diseased animals (P-value < 0.05), depression of MDA level, elevation of HO-1, GPx4 activity, and reduction of Cox2 and VEGF as compared with the diseased, AgNPs or zileuton-treated groups (P-value < 0.05). CONCLUSION The study showed that the combination of AgNPs and zileuton guards against, inflammation, apoptosis, and ferroptosis in PCO.
Collapse
Affiliation(s)
- Amira K Eltokhy
- Department of Medical Biochemistry, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Omnia Safwat El-Deeb
- Department of Medical Biochemistry, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman E Farghal
- Department of Clinical Pathology, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rowida Raafat Ibrahim
- Department of Medical Biochemistry, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rasha Elesawy
- Department of Pharmacology, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa Mahmoud Awad
- Department of Medical Physiology, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Radwa Ismail
- Department of Anatomy, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shaimaa M Motawea
- Department of Anatomy, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Doaa Shatat
- Department of Gynecology and Obstetrics, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasser Mostafa Hafez
- Internal Medicine Department, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hend Ahmed El Hanafy
- Department of Anatomy, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa Mohamed Atef
- Department of Medical Biochemistry, Tanta Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Chen X, Xiao Z, Cai Y, Pan Y. Reproductive hormone characteristics of obese Chinese patients with polycystic ovarian syndrome: a meta-analysis. Gynecol Endocrinol 2025; 41:2497854. [PMID: 40287875 DOI: 10.1080/09513590.2025.2497854] [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: 11/05/2024] [Revised: 03/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this analysis is to assess the effect of obesity on reproductive hormones in Chineses patients with polycystic ovarian syndrome (PCOS). Seven databases were searched. The Newcastle-Ottawa Scale (NOS) assessed the quality of included studies. A meta-analysis was performed using random-effects model. The means and standard deviations of the outcomes were synthesized as standardized mean differences (SMDs) with corresponding 95% confidence intervals (CIs). A total of 23 studies involving 4554 patients with PCOS were included. No significant differences in follicle-stimulating hormone (FSH) (p = 0.51), estradiol (E2) (p = 0.48), and prolactin (PRL) (p = 0.46) levels were found between obese and nonobese PCOS patients. However, obese PCOS patients had significantly lower levels of luteinizing hormone (LH) (p < 0.00001), LH/FSH (p = 0.001), progesterone (P) (p = 0.009), and anti-mullerian hormone (AMH) (p = 0.001). Conversely, they exhibited significantly higher testosterone (T) (p = 0.001) levels. Obese PCOS patients exhibited lower levels of LH, LH/FSH, P, and AMH, but higher T levels compared to nonobese PCOS patients, and no significant difference were observed in FSH, E2, and PRL levels in PCOS patients with and without obesity.
Collapse
Affiliation(s)
- Xiaolin Chen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuli Cai
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yizhuo Pan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Bashir SM, Ali SI, Rather MA, Sheikh WM, Singh H, Nabi SU, Ganie MA, Shafi M, Ul Haq Shah MZ, Bhat JI, Wani IA, Hassan S. Evaluating spironolactone monotherapy against combined treatment with metformin in rat PCOS model. Eur J Pharmacol 2025; 998:177516. [PMID: 40090535 DOI: 10.1016/j.ejphar.2025.177516] [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: 09/16/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a common gynecological disorder with multifactorial pathogenic risk factors. Combination therapy with metformin and thiazolidinedione derivatives is frequently used, but its synergistic effects have not been thoroughly evaluated. This study aims to compare the therapeutic efficacy of low-dose spironolactone (LDS) at 0.25 mg/kg for 28 days, metformin at 500 mg/kg for 28 days, and a combination of LDS and metformin, against a letrozole (1 mg/kg/day) and 0.5 % carboxymethylcellulose (CMC)-induced PCOS rat model. The study involved five groups of laboratory animals: Group I (Healthy control), Group IIa (Disease control), Group IIb (Metformin), Group IIc (LDS), and Group IId (Metformin + LDS). Therapeutic efficacy was evaluated based on phenotypic, hormonal, and genotypic determinants. Letrozole successfully induced PCOS in the animals, evidenced by elevated levels of Sex Hormone Binding Globulin (SHBG), Follicle Stimulating Hormone (FSH), and progesterone, as well as the presence of multiple ovarian cysts. Hierarchical Cluster Analysis indicated that LDS was superior to metformin and the combination therapy in ameliorating PCOS symptoms. The findings suggest that there is little to no benefit in adding metformin to LDS for the clinical management of PCOS. Although these results are from preclinical studies, further case-controlled, randomized placebo studies on a larger patient sample are necessary to confirm these findings in clinical settings.
Collapse
Affiliation(s)
- Showkeen Muzamil Bashir
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India.
| | - Sofi Imtiyaz Ali
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Muzafar Ahmad Rather
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Wajid Mohammad Sheikh
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates; Center for Biotechnology, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates
| | - Showkat Ul Nabi
- Division of Veterinary Clinical Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Mohd Ashraf Ganie
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu and Kashmir, 190001, India
| | - Majid Shafi
- Division of Veterinary Pathology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Mohd Zahoor Ul Haq Shah
- Laboratory of Endocrinology, Department of Bioscience, Barakatullah University Bhopal-462026, India
| | - Javeed Iqbal Bhat
- Biochemistry and Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, 190006, Jammu and Kashmir, India
| | - Imtiyaz Ahmad Wani
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu and Kashmir, 190001, India
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates; Center for Biotechnology, Khalifa University, Abu Dhabi, P.O 127788, United Arab Emirates.
| |
Collapse
|
4
|
Wang D, Li H, Wang Q, Liu Y. NAT10 mediated polycystic ovary syndrome through the ac4C modification of CXCL14. J Steroid Biochem Mol Biol 2025; 251:106767. [PMID: 40300667 DOI: 10.1016/j.jsbmb.2025.106767] [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: 08/16/2024] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder among women of reproductive age, underscoring the critical importance of investigating its regulatory mechanisms. N-Acetyltransferase 10 (NAT10) is a crucial enzyme involved in mRNA acetylation modification, mediating target genes expression through N4-acetylcytidine (ac4C) modification to regulate the biological function of various diseases. Nonetheless, the specific role of NAT10 in PCOS regulation remains undisclosed. Ac4C dot hybridization experiment was conducted to determine ac4C expression in PCOS tissues. RT-qPCR was employed to assess the expression levels of NAT10 and CXCL14 in PCOS tissues and KGN cells. Cells viability was assessed using the CCK-8 method, while cell proliferation capacity was evaluated through the colony formation assay and EDU assay. Flow cytometry analysis was utilized to measure the apoptosis rate. The ac4C modification level was determined by acrp-qPCR analysis. RIP and luciferase reporter experiments confirmed the target binding relationship. The rat experiments confirmed the specific regulatory role of NAT10 in polycystic ovary syndrome in vivo. This study highlighted reduced levels of NAT10 and ac4C in PCOS, where silencing NAT10 boosts KGN cell proliferation and suppresses apoptosis. Additionally, NAT10-mediated ac4C modification governed the chemokine CXCL14 expression. Our research unveiled that NAT10 modulated PCOS occurrence and progression by enhancing the CXCL14 mRNA stability through acetylation, presenting potential novel insights into the mechanisms of mRNA acetylation in PCOS.
Collapse
Affiliation(s)
- Ding Wang
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China.
| | - Hui Li
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| | - Qiaoling Wang
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| | - Yunxia Liu
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| |
Collapse
|
5
|
Garcia AV, Strangas Y. Cardiovascular Health in Women. Prim Care 2025; 52:317-328. [PMID: 40412909 DOI: 10.1016/j.pop.2025.01.005] [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
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the developed world. Although women generally have a lower prevalence, they have unique factors placing them at risk for CVD compared to their male counterparts. Studies show that prevention, early detection, and treatment can reduce downstream sequelae of CVD. However, sex and gender-specific recommendations are lagging and not always optimally disseminated to patients and providers. This article will explore sex and gender-based differences in cardiovascular burden of disease, risk factors, presentation, and natural history to aid in early identification and intervention.
Collapse
Affiliation(s)
- Aury V Garcia
- Department of Internal Medicine, Center for Family and Community Medicine; Columbia University Irving Medical Center/New York Presbyterian Hospital, 610 West 158th Street, New York, NY 10032, USA.
| | - Yorgos Strangas
- Department of Internal Medicine, Center for Family and Community Medicine; Columbia University Irving Medical Center/New York Presbyterian Hospital, 720 West 173rd Street Apartment 42, New York, NY 10032, USA
| |
Collapse
|
6
|
Moelle EC, Papps FA. Mental freedom and body acceptance by others as correlates of embodied well-being in women with polycystic ovary syndrome in Australia. Body Image 2025; 53:101894. [PMID: 40318525 DOI: 10.1016/j.bodyim.2025.101894] [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: 11/06/2024] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive-endocrinological-metabolic condition in women, causing bodies to diverge from cis-heteronormative feminine expectations. Greater body image concerns have been identified among women with PCOS, but further research is needed to identify drivers and mitigating factors. The ability to inhabit the body positively may be one such mitigating factor, and the developmental theory of embodiment is a novel framework allowing exploration of an integrated range of protective (and risk) sociocultural factors that may contribute to embodied well-being. This cross-sectional survey study therefore examined relationships among embodied well-being, severity of PCOS symptoms, body mass index (BMI), mental freedom (exposure to a social environment that facilitates an internalised critical stance towards oppressive social discourses, including femininity discourses on appearance and comportment), and perceived body acceptance by others in a sample of 140 women with PCOS, 18 years and over, living in Australia. Results of hierarchical multiple regression demonstrated that BMI and PCOS severity were negatively related to embodied well-being, but that when mental freedom and perceived body acceptance by others were added to the model, only PCOS severity, mental freedom, and body acceptance by others were significantly related to embodied well-being. Neither mental freedom nor body acceptance by others moderated the relationship between PCOS severity and BMI and embodied well-being. Results support BMI and PCOS severity as drivers of, and mental freedom and body acceptance by others as protective factors for, embodied well-being in women with PCOS, and have implications for interventions to support embodied well-being.
Collapse
Affiliation(s)
- Erika C Moelle
- Discipline of Psychological Science, ACAP University College, Australia.
| | - Fiona A Papps
- Discipline of Psychological Science, ACAP University College, Australia.
| |
Collapse
|
7
|
Ye Z, Cheng M, Lian W, Leng Y, Qin X, Wang Y, Zhou P, Liu X, Peng T, Wang R, He Y, Pan H, Zhao Y, Li R. GPX4 deficiency-induced ferroptosis drives endometrial epithelial fibrosis in polycystic ovary syndrome. Redox Biol 2025; 83:103615. [PMID: 40253746 PMCID: PMC12023900 DOI: 10.1016/j.redox.2025.103615] [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: 02/04/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025] Open
Abstract
The increased risk of infertility and endometrial lesions (such as endometrial hyperplasia or cancer) in polycystic ovary syndrome (PCOS) are closely associated with the lack of cyclical transformation in the endometrium. However, the underlying mechanisms remain incompletely understood. Though integrating single-cell RNA-sequencing, transcriptomics, and metabolomics analysis, we found that glutathione (GSH) metabolism disorder and the overactivation of ferroptosis, triggered by glutathione peroxidase 4 (GPX4) deficiency in endometrial epithelial cells, were the consequences of the prolonged endometrial proliferative phase in PCOS. This change may collectively contribute to some extent to decidualization failure. We further performed GSVA analysis and determined that the negative correlation between ferroptosis and fibrosis-related pathway was the most significant. Therefore, we first confirmed the presence of fibrosis in the proliferative endometrium of PCOS and PCOS-like mouse uteri. Additionally, by establishing endometrial organoids (EEOs) models and in vitro cell line models, we demonstrated that GPX4 deficiency contributed to extracellular matrix remodeling and excessive collagen deposition, via activating the TGF-β1/Smad2/3 pathway, which ultimately accelerated fibrosis. GSH intervention to the EEOs of PCOS could alleviate their fibrotic phenotypes at different stages. These findings may serve as a promising therapeutic target for PCOS-related endometrial dysfunction, as well as valuable strategies for improving PCOS-related adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Zhenhong Ye
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Ming Cheng
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Weisi Lian
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yueqi Leng
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xunsi Qin
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yue Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Ping Zhou
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xiyao Liu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Tianliu Peng
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Ruiqi Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yilei He
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Heng Pan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
8
|
Lu Y, Geng L, Zhou D, Sun Y, Xu H, Du X, Xu Q, Chen M. Toxic metals and trace elements, markers of inflammation, and hyperandrogenemia in women and testosterone deficiency in men: Associations and potential mediating factors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118352. [PMID: 40409193 DOI: 10.1016/j.ecoenv.2025.118352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025]
Abstract
There is evidence suggesting that toxic metal and trace element exposure disrupts androgen levels. However, specific effects of such exposure on hyperandrogenemia (HA) in women and testosterone deficiency (TD) in men remain unclear. The study investigated associations between toxic metal and trace element exposure and HA in women and TD in men, utilizing data from the National Health and Nutrition Examination Survey (NHANES) conducted between 2013 and 2016, and included 2205 women and 2621 men. Single-exposure analyses found lead (Pb) (adjusted odds ratio [aOR] 1.19, 95 % confidence interval [CI] 1.04-1.36) and cadmium (Cd) (aOR 1.20, 95 % CI 1.03-1.39) were positively associated with HA, especially strong in women over the age of 35. Manganese (Mn) was positively associated with TD (aOR 1.15, 95 % CI 1.03-1.28), especially strong in men aged between 45 and 50 years and obese men. A grouped weighted quantile sum (GWQS) model was performed to assess the effects of combined exposure, and found toxic metal mixture was positively associated with HA, particularly Cd (0.59) and Pb (0.40), and trace element mixture was positively associated with TD, particularly Mn (0.78). Mediation analyses demonstrated inflammation, particularly monocyte count and alkaline phosphatase (ALP) mediated 7.5-11.76 % of the associations. These findings provide insights to inform interventions for reducing exposure.
Collapse
Affiliation(s)
- Yingying Lu
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lulu Geng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Dan Zhou
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yan Sun
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Haoyi Xu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, China.
| | - Xinyi Du
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Qing Xu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing 210004, China.
| | - Miaoxin Chen
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
9
|
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
|
10
|
Ge H, Huang D, Tan L, Luo D, Zhou L, Liu H, Zhang Y, Liu D, Wu X, Wang L, Xiong L, Yang Y, Han TL, He C, Qi H. Metabolic Profiles of Pregnancy With Polycystic Ovary Syndrome: Insights into Maternal-Fetal Metabolic Communication. J Clin Endocrinol Metab 2025; 110:1524-1536. [PMID: 39880380 DOI: 10.1210/clinem/dgaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 01/31/2025]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) pregnancies are linked to metabolic disorders affecting maternal and fetal outcomes, with maternal metabolites differing from those in normal pregnancies. OBJECTIVE To investigate the metabolic communication at the maternal-fetal interface in PCOS pregnancies. DESIGN Placenta and umbilical cord serum were analyzed using gas chromatography-mass spectrometry. In-depth analysis was performed with clinical characteristics. SETTING Placenta and umbilical cord serum were analyzed using gas chromatography-mass spectrometry, alongside clinical characteristics. PARTICIPANTS Forty-five uncomplicated PCOS pregnancies and 50 normal pregnancies. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The metabolic characteristics at the maternal-fetal interface in PCOS pregnancies and the underlying mechanisms. RESULTS A total of 79 metabolites in the placenta and 25 in umbilical cord serum showed significant differences between PCOS and normal pregnancies. The 10 most significant placental metabolites were identified through receiver operating characteristic analysis, 9 of which correlated significantly with maternal serum testosterone levels. Lasso regression analysis identified 4 key placental metabolite combinations: gamma-aminobutyric acid, proline, glycine, and isoleucine, achieving an area under the curve of 93.24%. In umbilical cord serum, 6 metabolites differed significantly between PCOS and normal pregnancies, with the highest area under the curve reaching 76.07%; 5 of these metabolites showed significant correlations with maternal serum testosterone levels. Nine differential metabolites were shared between the placenta and umbilical cord serum, which also shared metabolic pathways, including ABC transporters and aminoacyl-tRNA biosynthesis, potentially influencing maternal-fetal interactions. CONCLUSION This study identifies the metabolomic profile and key pathways in maternal-fetal communication during PCOS pregnancies.
Collapse
Affiliation(s)
- Huisheng Ge
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Dongni Huang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lunbo Tan
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dan Luo
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Liu Zhou
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Hong Liu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Yilan Zhang
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Dandan Liu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Xixi Wu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Lulu Wang
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Liling Xiong
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610015, China
| | - Yang Yang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Chengjin He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing 401147, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Hanna A, Abbas H, Yassine F, AlBush A, Bilen M. Systematic review of gut microbiota composition, metabolic alterations, and the effects of treatments on PCOS and gut microbiota across human and animal studies. Front Microbiol 2025; 16:1549499. [PMID: 40438215 PMCID: PMC12116390 DOI: 10.3389/fmicb.2025.1549499] [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: 12/21/2024] [Accepted: 04/15/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Polycystic ovary syndrome (PCOS) is an endocrine disorder affecting around 12% of women globally, associated with infertility and various comorbidities. Emerging evidence suggests a crucial role of gut microbiota in PCOS pathophysiology, prompting research to investigate alterations in gut microbial composition in patients with PCOS. Methods This systematic review aims to analyze human and animal studies that compare gut microbiota composition, gut-derived metabolites, and treatment interventions in PCOS patients versus healthy controls. A comprehensive literature search was conducted using PubMed, Scopus, and Web of Science, yielding studies examining gut microbiota, metabolomic shifts, and treatment responses in PCOS models and human populations. Results Our analysis revealed decreases in alpha diversity in PCOS patients, with more pronounced changes in beta diversity in animal models. Specific bacterial taxa, such as Bacteroides vulgatus, Escherichia-Shigella and Lactobacillus, showed implication in PCOS pathogenesis, suggesting potential microbial markers. Furthermore, discrepancies between human and animal studies show the need for humanized mouse models to bridge this gap. Interventions like probiotics and fecal microbiota transplantation (FMT) showed varying levels of efficacy, with FMT emerging as a more promising but invasive option, offering live bacteriotherapy as a potential therapeutic alternative. Alterations in gut-derived metabolites, including short-chain fatty acids and bile acids, highlighted the multifaceted nature of PCOS, with implications extending to metabolic, hormonal, and gut-brain axis disruptions. Discussion In conclusion, PCOS exhibits complex interactions between gut microbiota and metabolic pathways, necessitating further research with standardized methods and larger sample sizes to elucidate the microbiome's role in PCOS.
Collapse
Affiliation(s)
- Aya Hanna
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Abbas
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fayez Yassine
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Alia AlBush
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Melhem Bilen
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Centre for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- World Health Organization (WHO) Collaborating Centre for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| |
Collapse
|
12
|
Bezdíček J, Sekaninová J, Janků M, Makarevič A, Luhová L, Dujíčková L, Petřivalský M. Reactive oxygen and nitrogen species: multifaceted regulators of ovarian activity†. Biol Reprod 2025; 112:789-806. [PMID: 39936599 PMCID: PMC12078082 DOI: 10.1093/biolre/ioaf032] [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: 05/02/2024] [Revised: 09/10/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
Reactive oxygen and nitrogen species are essential components of diverse intracellular signaling pathways. In addition to their involvement in apoptosis, reactive oxygen and nitrogen species are crucial in the regulation of multiple developmental and physiological processes. This review aims to summarize their role in the regulation of key ovarian stages: ovulation, maturation and postovulatory ageing of the oocyte, and the formation and regression of the corpus luteum. At the cellular level, a mild increase in reactive oxygen and nitrogen species is associated with the initiation of a number of regulatory mechanisms, which might be suppressed by increased activity of the antioxidant system. Moreover, a mild increase in reactive oxygen and nitrogen species has been linked to the control of mitochondrial biogenesis and abundance in response to increased cellular energy demands. Thus, reactive oxygen and nitrogen species should also be perceived in terms of their positive role in cellular signaling. On the other hand, an uncontrolled increase in reactive oxygen species production or strong down-regulation of the antioxidant system results in oxidative stress and damage of cellular components associated with ovarian pathologies and ageing. Similarly, the disturbance of signaling functions of reactive nitrogen species caused by dysregulation of nitric oxide production by nitric oxide synthases in ovarian tissues interferes with the proper regulation of physiological processes in the ovary.
Collapse
Affiliation(s)
- Jiří Bezdíček
- Department of Zoology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Jana Sekaninová
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Martina Janků
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Alexander Makarevič
- National Agricultural and Food Centre, Research Institute for Animal Production Nitra, Lužianky-near-Nitra, Slovak Republic
| | - Lenka Luhová
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Linda Dujíčková
- National Agricultural and Food Centre, Research Institute for Animal Production Nitra, Lužianky-near-Nitra, Slovak Republic
| | - Marek Petřivalský
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| |
Collapse
|
13
|
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
|
14
|
Morin-Papunen L, Pelkonen S, Piltonen T. Polycystic ovary syndrome: What to say when asked about the chance of pregnancy. Acta Obstet Gynecol Scand 2025. [PMID: 40329712 DOI: 10.1111/aogs.15149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Affiliation(s)
- Laure Morin-Papunen
- Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Sari Pelkonen
- Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Oulu University Hospital, Oulu, Finland
| | - Terhi Piltonen
- Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
15
|
Geng L, Yang X, Sun J, Ran X, Zhou D, Ye M, Wen L, Wang R, Chen M. Gut Microbiota Modulation by Inulin Improves Metabolism and Ovarian Function in Polycystic Ovary Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412558. [PMID: 40192074 PMCID: PMC12120758 DOI: 10.1002/advs.202412558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/27/2025] [Indexed: 06/01/2025]
Abstract
The management of metabolic disorder associated with polycystic ovary syndrome (PCOS) has been suggested as an effective approach to improve PCOS which is highly involved with gut microbiota, while the underlying mechanism is unclear. Here, we investigated the role of inulin, a gut microbiota regulator, in the alleviation of PCOS. Our findings showed that inulin treatment significantly improved hyperandrogenism and glucolipid metabolism in both PCOS cohort and mice. Consistent with the cohort, inulin increased the abundance of microbial co-abundance group (CAG) 12 in PCOS mice, including Bifidobacterium species and other short-chain fatty acids (SCFAs)-producers. We further verified the enhancement of SCFAs biosynthesis capacity and fecal SCFAs content by inulin. Moreover, inulin decreased lipopolysaccharide-binding protein (LBP) and ameliorated ovarian inflammation in PCOS mice, whereas intraperitoneal lipopolysaccharide (LPS) administration reversed the protective effects of inulin. Furthermore, fecal microbiota transplantation (FMT) from inulin-treated patients with PCOS enhanced insulin sensitivity, improved lipid accumulation and thermogenesis, reduced hyperandrogenism and ovarian inflammatory response in antibiotic-treated mice. Collectively, these findings revealed that gut microbiota mediates the beneficial effects of inulin on metabolic disorder and ovarian dysfunction in PCOS. Therefore, modulating gut microbiota represents a promising therapeutic strategy for PCOS.
Collapse
Affiliation(s)
- Lulu Geng
- Centre for Assisted ReproductionShanghai Key Laboratory of Maternal‐Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Xin Yang
- Shanghai Innovation Center of TCM Health ServiceShanghai University of Traditional Chinese MedicineShanghai201203China
- Section of Endocrinology, Internal MedicineSchool of MedicineYale UniversityNew HavenCT06520USA
| | - Jiani Sun
- Centre for Assisted ReproductionShanghai Key Laboratory of Maternal‐Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Ximing Ran
- Department of Biostatistics and BioinformaticsRollins School of Public HealthEmory UniversityAtlantaGA30322USA
| | - Dan Zhou
- Centre for Assisted ReproductionShanghai Key Laboratory of Maternal‐Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Mingming Ye
- Centre for Assisted ReproductionShanghai Key Laboratory of Maternal‐Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Li Wen
- Section of Endocrinology, Internal MedicineSchool of MedicineYale UniversityNew HavenCT06520USA
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health ServiceShanghai University of Traditional Chinese MedicineShanghai201203China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Miaoxin Chen
- Centre for Assisted ReproductionShanghai Key Laboratory of Maternal‐Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| |
Collapse
|
16
|
Li Y, Lin X, Zou K, Du J, Li Q, Zhong L, Jiang S. Blood biochemical landscape and new insights into clinical decision-making for polycystic ovary syndrome in Chinese women: a prospective cohort study. Front Endocrinol (Lausanne) 2025; 16:1534733. [PMID: 40375948 PMCID: PMC12078145 DOI: 10.3389/fendo.2025.1534733] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction The Polycystic ovary syndrome (PCOS), a prevalent endocrine disorder affecting women's reproductive and metabolic health, faces diagnostic challenges due to heterogeneous clinical presentations and the absence of reliable biomarkers. This study investigates the role of Glucosaminyl (N-acetyl) transferase 2 (GCNT2) in modulating sex hormone-binding globulin (SHBG) and its potential as a therapeutic target in PCOS pathophysiology. Methods A prospective cohort of 103 PCOS patients treated with oral contraceptives (2021-2024) was established. Bidirectional Mendelian randomization (MR) was employed to assess genetic associations and causal relationships between PCOS and SHBG. Molecular docking studies evaluated cryptotanshinone's binding affinity to key proteins (COL1A1, COL4A2, COL6A2) in the PI3K/Akt pathway. GCNT2's regulatory effects on collagen synthesis and extracellular matrix pathways. Pharmacokinetic profiling validated therapeutic viability. Results Bidirectional MR revealed significant genetic associations (P < 0.001) and causal links between PCOS and SHBG, implicating GCNT2 as a key modulator. Cryptotanshinone exhibited strong binding affinity to PI3K/Akt signaling pathway proteins and favorable pharmacokinetic properties. Enrichment analyses highlighted GCNT2's role in collagen biosynthesis (FDR < 0.05) and extracellular matrix regulation. Discussion This study identifies GCNT2 as a critical mediator of PCOS pathophysiology through SHBG modulation and collagen remodeling. Cryptotanshinone emerges as a promising therapeutic candidate, targeting PI3K/Akt signaling pathway with high specificity. These findings advance the understanding of PCOS mechanisms and provide a foundation for biomarker-driven diagnostics and precision therapeutics. Further validation in clinical trials is warranted to translate these insights into practice.
Collapse
Affiliation(s)
- Yutong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Xiufeng Lin
- Reproductive Center, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Ke Zou
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jing Du
- Reproductive Center, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Qingni Li
- Reproductive Center, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Linkun Zhong
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Shan Jiang
- Reproductive Center, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| |
Collapse
|
17
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of PCOS by developmental programming and germline transmission. Trends Endocrinol Metab 2025; 36:472-481. [PMID: 39732517 DOI: 10.1016/j.tem.2024.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder, affecting approximately 11-13% of women of reproductive age. Women with PCOS experience a higher prevalence of infertility, pregnancy complications, and cardiometabolic disorders such as obesity, insulin resistance, and type 2 diabetes mellitus. Furthermore, psychiatric comorbidities, including depression and anxiety, significantly impact the quality of life in this population. Although obesity exacerbates these health risks, the exact etiology and pathophysiology of PCOS remain complex and only partially understood. Emerging research suggests potential transgenerational inheritance through genetic and epigenetic mechanisms, highlighting the possibility of PCOS-related risks affecting subsequent generations, including sons. This review synthesizes recent findings on PCOS inheritance patterns and underscores areas for future clinical and research exploration.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
18
|
Racine C, Fraissinet F, Tolu S, Pereira T, Gil S, Badel A, Bailbé D, Fève B, Movassat J, Cate R, di Clemente N. A blocking antibody against anti-Müllerian hormone restores ovulation and normal androgen levels in a spontaneous rat model of polycystic ovary syndrome. EBioMedicine 2025; 115:105716. [PMID: 40252252 PMCID: PMC12032919 DOI: 10.1016/j.ebiom.2025.105716] [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: 09/12/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Polycystic Ovary Syndrome (PCOS), the leading cause of infertility worldwide, is characterised by oligo-anovulation, hyperandrogenism, polycystic ovarian morphology and high Anti-Müllerian hormone (AMH) levels, associated with severe metabolic disturbances. However, the role of AMH in the physiopathology of this syndrome remains poorly understood and strategies to block its effects have never been investigated in animal models of PCOS. METHODS We used Western-blotting, ELISA and gene reporter approaches to evaluate the blocking efficacy, interspecificity and mechanism of action of an antibody against human AMH, Mab22A2. Then, we investigated the ability of a rat version of Mab22A2, rMab22A2, to alleviate reproductive dysfunction in Goto-Kakizaki (GK) rats, which spontaneously exhibit all the features of women with PCOS. FINDINGS We showed that Mab22A2 was interspecific, did not prevent AMH from binding to its receptor and was able to block the effects of AMH in gonadal cell lines. In addition, treatment of anovulatory GK rats with rMab22A2 reduced their bioavailable serum AMH levels and normalised their androgen concentrations. Finally, this treatment also induced ovulation in 84% of the rats and resulted in 66% of pregnancies. INTERPRETATION Our results show that AMH is a major driver of reproductive and hormonal dysfunction in PCOS and provide proof of concept that a blocking antibody against AMH can reverse the major reproductive dysfunction observed in PCOS, opening up promising avenues for the treatment of patients with PCOS. FUNDING Inserm, Sorbonne University, Inserm Transfert, the French Endocrine Society and the Medical Research Foundation (grant agreement n°EQU201903007868).
Collapse
Affiliation(s)
- Chrystèle Racine
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Université Paris Cité, Paris 75013, France
| | - François Fraissinet
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | - Stefania Tolu
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Tony Pereira
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | | | - Anne Badel
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Danielle Bailbé
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Bruno Fève
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Department of Endocrinology, Hôpital Saint-Antoine, APHP, Paris 75012, France
| | | | - Richard Cate
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Nathalie di Clemente
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France.
| |
Collapse
|
19
|
Whooten RC, Rifas-Shiman SL, Perng W, Chavarro JE, Oken E, Hivert MF. Physical activity may attenuate the association of early adolescent adiposity with PCOS assessed in later adolescence: findings from a prospective cohort, Project Viva. J Pediatr Adolesc Gynecol 2025:S1083-3188(25)00262-1. [PMID: 40316007 DOI: 10.1016/j.jpag.2025.04.011] [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: 10/01/2024] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVE To assess how moderate-vigorous physical activity (MVPA) may modify the association of early adolescent adiposity with late adolescent polycystic ovary syndrome (PCOS). METHODS Within the Project Viva prospective cohort, we assessed the association of body mass index z-score (BMIz) at early adolescence (mean age 13.1 years) with later PCOS (self-reported diagnosis or oligo-anovulation with clinical/biochemical hyperandrogenism) at mean age 17.7 years. We stratified our analyses by tertile of actigraphy-assessed moderate-vigorous physical activity (MVPA) during early adolescence . In multivariable logistic regression models, we adjusted for maternal education and PCOS, child race and ethnicity, and child health behaviors. RESULTS Among 341 female adolescents, n=47 (14%) met criteria for PCOS in late adolescence. In early adolescence (∼13 years old), mean (SD) BMIz was 0.32 (1.10) units and mean (SD) MVPA was 27.1 (17.9) min/day. After adjusting for covariates, early adolescent BMIz was associated with later PCOS (OR=1.72 95%CI=1.15, 2.56 per unit increase in BMIz). In stratified analyses, the association of early adolescent BMIz with later PCOS was apparent for those in the lowest tertile of MVPA (OR 3.22; 95% CI 1.25, 8.31) however was weaker among those more active (MVPA tertile 2 OR = 1.65; 95% CI 0.74, 3.67 and tertile 3 OR=1.34; 95% CI=0.63, 2.86, respectively).The interaction term p-value was non-significant (p=0.36). CONCLUSION Our findings suggest that the detrimental effect of adiposity in relation to PCOS risk is most apparent only among those with lower MVPA. Maintaining adequate MVPA during the early teen years may mitigate PCOS risk associated with excess adiposity.
Collapse
Affiliation(s)
- Rachel C Whooten
- Division of Pediatric Endocrinology, Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA; Division of General Academic, Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA.
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Wei Perng
- Department of Epidemiology, Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jorge E Chavarro
- Department of Nutrition, T. H. Chan Harvard School of Public Health, Boston, Massachusetts, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA; Department of Nutrition, T. H. Chan Harvard School of Public Health, Boston, Massachusetts, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA; Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
20
|
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
|
21
|
Ajorlouie Z, Moshkian P, Baghdadi G, Amiri R, Biglari F, Rahimlou M. The association between the Mediterranean Diet and the Prime Diet Quality Score and polycystic ovary syndrome: a case control study. BMC Nutr 2025; 11:80. [PMID: 40241157 PMCID: PMC12001616 DOI: 10.1186/s40795-025-01067-5] [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: 09/07/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. This study aims to investigate the association between adherence to the Mediterranean Diet (Med-Diet) and the Prime Diet Quality Score (PDQS) and the risk of PCOS. METHOD This case-control study included 472 women aged 18-45, with 180 PCOS cases and 292 controls. PCOS diagnosis was based on the Rotterdam criteria. Dietary intake was assessed using a validated food frequency questionnaire, and adherence to the Med-Diet and PDQS was calculated. Statistical analyses included logistic regression to examine associations between diet quality and PCOS. RESULTS Higher adherence to both the Med-Diet and PDQS was significantly associated with lower odds of PCOS. Participants in the highest quartile of the Med-Diet score had a 41% reduced risk of PCOS in the crude model (OR = 0.59, 95% CI: 0.48-0.67) and a 32% reduced risk in the fully adjusted model (OR = 0.68, 95% CI: 0.57-0.79), after adjusting for potential confounders, including age, body mass index (BMI), physical activity, and total energy intake. Similarly, those in the highest PDQS quartile showed a 53% reduced risk in the crude model (OR = 0.47, 95% CI: 0.35-0.56) and a 43% reduced risk in the fully adjusted model (OR = 0.57, 95% CI: 0.44-0.68), accounting for the same confounders. CONCLUSION The findings suggest that higher adherence to the Med-Diet and PDQS is associated with a reduced risk of developing PCOS. Further research is warranted to explore the underlying biological mechanisms and to establish causality through prospective cohort studies and randomized controlled trials.
Collapse
Affiliation(s)
- Zeinab Ajorlouie
- Department of Midwifery, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Paniz Moshkian
- Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazal Baghdadi
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Roksaneh Amiri
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fereshteh Biglari
- Department of educational management, Zanjan university of medical science, 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 Sciences, Zanjan, Iran.
| |
Collapse
|
22
|
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
|
23
|
Gao Q, Qian X, Zhang J, Zhang F, Xu X. Body image disturbances in women of childbearing age with polycystic ovary syndrome: a cross-sectional study. BMC Womens Health 2025; 25:186. [PMID: 40241131 PMCID: PMC12001658 DOI: 10.1186/s12905-025-03693-4] [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: 09/12/2023] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
DISIGN This is a cross-sectional study. OBJECTIVES This study aimed to identify the risk factors associated with body image disturbances in patients with polycystic ovary syndrome (PCOS). METHODS This study included married women diagnosed with PCOS who required fertility assistance. Participants were recruited from the reproductive centers of three tertiary hospitals in Jiangsu Province between December 2022-June 2023. We collected 333 patient data through an online questionnaire, body image was assessed by the Chinese version of Body Image Disturbance Questionnaire (BIDQ). The demographic information and clinical data of patients were collected. At the same time, the relevant information of sleep quality, personality traits, intimate relationship, family care and psychological resilience of the patients were collected through various scales. Univariate analysis and a generalized linear model were employed to analyze the factors affecting body image disturbances in women with PCOS. RESULTS This cross-sectional study included 333 women of childbearing age with PCOS. Univariate analysis revealed that body image was associated with factors such as the body mass index (BMI), occupation, menstrual disturbances, a family history of PCOS, hair loss, acne, hirsutism, scores on the Family Apgar Index, psychological resilience levels, sleep quality, intimate relationship, and personality traits. The generalized linear model demonstrated that occupation, BMI, hair loss, acne, hirsutism, intimate relationship, family care, and psychological resilience were independent risk factors for body image issues. CONCLUSION The influencing factors of body image disturbance in patients with polycystic ovary syndrome of childbearing age come from many aspects, including occupation, acne, hair loss, hirsutism, BMI, intimate relationship, family care and psychological resilience. Consequently, medical professionals in obstetrics and gynecology should recognize the body image concerns of women with PCOS and implement specific interventions to support these women during their childbearing years.
Collapse
Affiliation(s)
- Qian Gao
- Department of Nursing, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
- School of Nursing and Rehabilitation, Nantong University, Nantong, Jiangsu, 226001, China
| | - Xuan Qian
- Department of Nursing, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Jie Zhang
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Feng Zhang
- School of Nursing and Rehabilitation, Nantong University, Nantong, Jiangsu, 226001, China.
| | - Xujuan Xu
- Department of Nursing, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
24
|
Cotellessa L, Sobrino V, Silva MSB, Delit M, Maitre H, Caron E, Ternier G, da Silva Lima N, Lhomme T, Giton F, Sorrentino A, Carraresi L, Di Nardo G, Nogueiras R, Tena-Sempere M, Prevot V, Giacobini P. Preventing and correcting polycystic ovary syndrome by targeting anti-Müllerian hormone signaling in minipuberty and adulthood in mice. Cell Metab 2025:S1550-4131(25)00116-0. [PMID: 40220763 DOI: 10.1016/j.cmet.2025.03.013] [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: 09/23/2024] [Revised: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrinopathy in women, causes significant reproductive and metabolic comorbidities, with no current cure. Gestational androgen and anti-Müllerian hormone (AMH) excess are linked to PCOS, and prenatal aberrant exposure to these hormones induces PCOS-like traits in animal models. However, whether the AMH effects on PCOS programming could extend to early postnatal life remains unknown. Clinical observations showed higher AMH levels during minipuberty in infants of mothers with PCOS, but whether this contributes to PCOS development is uncertain. Here, we show that exposure to high AMH levels during minipuberty in mice causes PCOS-like reproductive and metabolic defects in both sexes. A neutralizing antibody targeting AMH receptor 2 (AMHR2) prevented these defects when administered during minipuberty and alleviated symptoms when given in adulthood. These findings highlight the causal role of elevated AMH in PCOS and suggest AMHR2-targeting therapy as a potential preventive or curative approach.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Veronica Sobrino
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC) and Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Mauro S B Silva
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Maxime Delit
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Hélène Maitre
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Emilie Caron
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaëtan Ternier
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Natalia da Silva Lima
- CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Tori Lhomme
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Frank Giton
- IMRB - Inserm U955, Faculté de Santé, Créteil, France
| | - Andrea Sorrentino
- DIVAL Toscana Srl, Sesto Fiorentino, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Carraresi
- DIVAL Toscana Srl, Sesto Fiorentino, Florence, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giovanna Di Nardo
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Manuel Tena-Sempere
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC) and Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Paolo Giacobini
- University Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France.
| |
Collapse
|
25
|
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
|
26
|
Liu Y, Dong Y, Jiang Y, Han S, Liu X, Xu X, Zhu A, Zhao Z, Gao Y, Zou Y, Zhang C, Bian Y, Zhang Y, Liu J, Zhao S, Zhao H, Chen ZJ. Caloric restriction prevents inheritance of polycystic ovary syndrome through oocyte-mediated DNA methylation reprogramming. Cell Metab 2025; 37:920-935.e6. [PMID: 39986273 DOI: 10.1016/j.cmet.2025.01.014] [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: 05/22/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/24/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent metabolic and reproductive endocrine disorder with strong heritability. However, the independent role of oocytes in mediating this heritability remains unclear. Utilizing in vitro fertilization-embryo transfer and surrogacy, we demonstrated that oocytes from androgen-exposed mice (F1) transmitted PCOS-like traits to F2 and F3 generations. Notably, caloric restriction (CR) in F1 or F2 effectively prevented this transmission by restoring disrupted DNA methylation in oocyte genes related to insulin secretion and AMPK signaling pathways. Further detection in adult tissues of offspring revealed dysregulated DNA methylation and expression of those genes (e.g., Adcy3, Gnas, and Srebf1) were reversed by maternal CR. Moreover, similar benefits of CR were observed in aberrant embryonic methylome of women with PCOS. These findings elucidate the essential role of CR in preventing PCOS transmission via methylation reprogramming, emphasizing the importance of preconception metabolic management for women with PCOS.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Center for Reproductive Medicine, Gusu School, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing 212028, Jiangsu, China
| | - Yi Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yonghui Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Shan Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Xin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Aiqing Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Zihe Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuan Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yang Zou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Chuanxin Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuehong Bian
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuqing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Jiang Liu
- Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Shigang Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China.
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Center for Reproductive Medicine, Gusu School, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing 212028, Jiangsu, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China.
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200025, China; Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
27
|
Pereira JD, Magalhães FMV, Tameirão FMS, Soriani FM, de O S Jorge KT, Reis FM, Cândido AL, Comim FV, Gomes KB. The possible regulatory role of miRNA-30c-5p, miRNA-545-3p and miRNA-125a-5p in women with polycystic ovary syndrome: A case-control study and signaling pathways. Mol Cell Endocrinol 2025; 599:112492. [PMID: 39952313 DOI: 10.1016/j.mce.2025.112492] [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: 11/18/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Polycystic Ovary Syndrome (PCOS) is one of the most common endocrinopathy in women of reproductive age. MicroRNA (miRNAs) are small non-coding RNAs related to the control of gene expression in biological fluids. Our study analyzed the expression of miRNAs related to inflammation in individuals with PCOS compared to controls. METHODS Twenty patients with PCOS and 20 controls, matched by body mass index and age, were included in the study. The miRNAs evaluated were miRNA-30c-5p; miRNA-545-3p and miRNA-125a-5p. RESULTS The expression of the miRNAs was similar between the two groups. A positive correlation was observed between the expression of miRNA-125a-5p and LDLc levels only in the PCOS group. Subsequent analysis of biological pathways showed that miRNA-125a -5p is significantly involved in the regulation of SREBP/SREBF pathways of cholesterol biosynthesis, glycolysis, insulin receptor signaling, oxidative stress-induced senescence and estrogen-dependent gene expression. CONCLUSION The results suggest that the miRNA-125a-5p shows a potential implication to the regulation of lipid biosynthesis and LDL-c levels in PCOS women.
Collapse
Affiliation(s)
- Jessica D Pereira
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda M V Magalhães
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabiana M S Tameirão
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Frederico M Soriani
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina T de O S Jorge
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando M Reis
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Lúcia Cândido
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio V Comim
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina B Gomes
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
28
|
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
|
29
|
Eriksson G, Li C, Sparovec TG, Dekanski A, Torstensson S, Risal S, Ohlsson C, Hirschberg AL, Petropoulos S, Deng Q, Stener-Victorin E. Single-cell profiling of the human endometrium in polycystic ovary syndrome. Nat Med 2025:10.1038/s41591-025-03592-z. [PMID: 40114027 DOI: 10.1038/s41591-025-03592-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Polycystic ovary syndrome (PCOS) has a negative effect on the receptivity of the endometrium to embryo implantation and increases the risk of miscarriage and endometrial cancer. The cellular and molecular heterogeneity of the endometrium in women with PCOS has not been well studied. Our study presents a comprehensive cellular atlas of the endometrium during the proliferative phase in women with PCOS characterized by overweight and obesity, hyperandrogenism and insulin resistance compared with controls of similar age, weight and body mass index. Analysis of 247,791 isolated endometrial nuclei from 27 biopsies (5 controls and 12 PCOS cases at baseline and 7 after 16 weeks of metformin and 3 after lifestyle intervention) revealed cell-type-specific disease signatures and variations in cellular composition and localization. Samples taken after 16 weeks of metformin treatment and lifestyle management showed extensive recovery of disease-specific endometrial signatures. We linked the specific role of each cell type to clinical features such as hyperandrogenism and insulin resistance, and specific cell types to risk of endometrial and metabolic disease. In addition, potential therapeutic targets such as integrin inhibitors were identified and the role of metformin in restoring endometrial health in patients with PCOS was highlighted. Our findings lay the groundwork to significantly advance the understanding of PCOS-specific endometrial dysfunction for future targeted therapies.
Collapse
Affiliation(s)
- Gustaw Eriksson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Congru Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tina Gorsek Sparovec
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anja Dekanski
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sara Torstensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sanjiv Risal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics/Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Claes Ohlsson
- Department of Drug Treatment, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sophie Petropoulos
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics/Gynecology, Karolinska Institutet, Stockholm, Sweden.
- Faculty of Medicine, Département de Médecine, Université de Montréal, Montréal, Québec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montréal, Québec, Canada.
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | | |
Collapse
|
30
|
Wang T, Chen X, Li C. SRC involves in lysosomal function and regulates ferroptosis in polycystic ovary syndrome. J Ovarian Res 2025; 18:42. [PMID: 40045372 PMCID: PMC11881414 DOI: 10.1186/s13048-025-01637-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: 12/22/2024] [Accepted: 02/23/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The pathogenesis of polycystic ovary syndrome (PCOS) is still unknown, so finding the molecular mechanisms of pathogenesis is crucial in PCOS. METHODS The GSE34526 dataset from the Gene Expression Omnibus (GEO) database was used to screen biomarkers in this study. KEGG enrichment analysis of GSE34526 was performed using Gene Set Enrichment Analysis (GSEA). The differentially expressed genes(DEGs) were screened and analyzed for lysosome-related genes. Subsequently, further KEGG and GO analyses were performed, and it was found that it was enriched in the ferroptosis pathway, and then the ferroptosis-related differential genes were obtained. The genes at the core position were obtained by the Protein-Protein Interaction(PPI) network. We then focused our attention on SRC and verified the differential expression of SRC in ovarian tissues of hyperandrogenemic, hyperlipemic and control groups, as well as the differences in conception rate and litter rate of each group by rat test. RESULTS GSEA analysis of the gene dataset GSE34526 revealed that LYSOSOME was significantly enriched in the PCOS group. There were 188 lysosome-related differentially expressed genes(LRDEGs) in granulosa cells from patients with PCOS, and 41 ferroptosis-related differentially expressed genes(FRDEGs). It was found that six of these genes, SRC, NCF2, SLC2A8, FTL, SLC2A6, SLC3A2, were present in all three datasets. SRC was the top ranked gene in the PPI network of FRDEGs.As verified by the rat model, the expression of SRC in the ovarian tissues of the hyperandrogenemic group was significantly higher than that of the control group (P=0.004) and the hyperlipemic group (P=0.002). CONCLUSION SRC, as an important gene involved in lysosomal function and regulating ferroptosis, is expected to be a potential target for PCOS.
Collapse
Affiliation(s)
- Tianmei Wang
- Department of Gynecology, First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400016, P.R. China
| | - Xin Chen
- Department of Gynecology and Obstetrics, The 958th Army Hospital of the Chinese People'S Liberation Army, Jiangbei District, Chongqing, 400000, P.R. China
| | - Cong Li
- Department of Gynecology, First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400016, P.R. China.
| |
Collapse
|
31
|
Mittal R, Prasad K, Lemos JRN, Arevalo G, Hirani K. Unveiling Gestational Diabetes: An Overview of Pathophysiology and Management. Int J Mol Sci 2025; 26:2320. [PMID: 40076938 PMCID: PMC11900321 DOI: 10.3390/ijms26052320] [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: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by an inadequate pancreatic β-cell response to pregnancy-induced insulin resistance, resulting in hyperglycemia. The pathophysiology involves reduced incretin hormone secretion and signaling, specifically decreased glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), impairing insulinotropic effects. Pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), impair insulin receptor substrate-1 (IRS-1) phosphorylation, disrupting insulin-mediated glucose uptake. β-cell dysfunction in GDM is associated with decreased pancreatic duodenal homeobox 1 (PDX1) expression, increased endoplasmic reticulum stress markers (CHOP, GRP78), and mitochondrial dysfunction leading to impaired ATP production and reduced glucose-stimulated insulin secretion. Excessive gestational weight gain exacerbates insulin resistance through hyperleptinemia, which downregulates insulin receptor expression via JAK/STAT signaling. Additionally, hypoadiponectinemia decreases AMP-activated protein kinase (AMPK) activation in skeletal muscle, impairing GLUT4 translocation. Placental hormones such as human placental lactogen (hPL) induce lipolysis, increasing circulating free fatty acids which activate protein kinase C, inhibiting insulin signaling. Placental 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) overactivity elevates cortisol levels, which activate glucocorticoid receptors to further reduce insulin sensitivity. GDM diagnostic thresholds (≥92 mg/dL fasting, ≥153 mg/dL post-load) are lower than type 2 diabetes to prevent fetal hyperinsulinemia and macrosomia. Management strategies focus on lifestyle modifications, including dietary carbohydrate restriction and exercise. Pharmacological interventions, such as insulin or metformin, aim to restore AMPK signaling and reduce hepatic glucose output. Emerging therapies, such as glucagon-like peptide-1 receptor (GLP-1R) agonists, show potential in improving glycemic control and reducing inflammation. A mechanistic understanding of GDM pathophysiology is essential for developing targeted therapeutic strategies to prevent both adverse pregnancy outcomes and the progression to overt diabetes in affected women.
Collapse
Affiliation(s)
| | | | | | | | - Khemraj Hirani
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (K.P.); (J.R.N.L.); (G.A.)
| |
Collapse
|
32
|
Yang Y, Liu L, Hu N, Huo H, Yang X, Wang F. Analysis of risk factors for depression and anxiety in women with polycystic ovary syndrome. Front Glob Womens Health 2025; 6:1520641. [PMID: 40098730 PMCID: PMC11911363 DOI: 10.3389/fgwh.2025.1520641] [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: 10/31/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is one of the most common reproductive endocrine disorders among women of reproductive age, often accompanied by a series of symptoms such as hirsutism, hair loss, menstrual disorders and obesity, resulting in an increasing risk of depression and anxiety in such patients. Methods A total of 413 patients in the Reproductive Medicine Center of the Second Hospital of Lanzhou University from June 2021 to June 2023 were enrolled. We collected sociodemographic information and lifestyle-related factors using a structured questionnaire. Patient Health Questionnaire (PHQ-9) and Generalized Anxiety Disorder Scale (GAD-7) were used to evaluate the psychological status of the subjects. Sleep-related variables were assessed using the Pittsburgh Sleep Quality Index (PSQI), and metabolic measures were collected from patients' medical records. Results Compared with the control group, PCOS patients were younger, the average age was (27.39 ± 3.48) years old, and the BMI value was higher, the difference was statistically significant (p < 0.05). The proportions of depression and anxiety in PCOS patients were 47.7% and 39.9%, respectively. In PCOS patients with depressive anxiety symptoms, the proportions of mild, moderate, moderately severe and severe depression were 31.6%, 12.4%, 1.6% and 2.1%, respectively. The proportions of mild, moderate, moderately severe and severe anxiety were 30.6%, 6.2%, 1.0% and 2.1%, respectively. Depression was significantly associated with serum free triiodothyronine (FT3) OR (95% CI) = 3.33 (1.30-8.55), sleep duration 4.99 (1.45-17.23) and daytime dysfunction 8.24 (3.53-19.22). Anxiety was significantly associated with daytime dysfunction OR (95% CI) = 3.45 (1.78-6.70). No association was found between mental health and other metabolic characteristics in PCOS patients (p > 0.05). Conclusion According to the results of the current study, a high proportion of women with PCOS have mental health disorders, and there is a significant correlation between mental health disorders and sleep conditions.
Collapse
Affiliation(s)
- Yanting Yang
- Department of Obstetrics and Gynecology, Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lin Liu
- Department of Obstetrics and Gynecology, Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ning Hu
- Department of Obstetrics and Gynecology, Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Huyan Huo
- Department of Obstetrics and Gynecology, Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xin Yang
- Department of Reproductive Medicine, Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Fang Wang
- Department of Reproductive Medicine, Second Hospital, Lanzhou University, Lanzhou, China
| |
Collapse
|
33
|
Ollila MM, Hautakoski A, Arffman M, Morin-Papunen L, Koivikko M, Ebeling T, Sund R, Piltonen T. Diabetic complications in women with type 2 diabetes and polycystic ovary syndrome. Eur J Endocrinol 2025; 192:202-209. [PMID: 39980352 DOI: 10.1093/ejendo/lvaf026] [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: 08/21/2024] [Revised: 12/06/2024] [Accepted: 02/19/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE To investigate whether increased prevalence of type 2 diabetes (T2DM) in women with polycystic ovary syndrome (PCOS) translates into increased risk of diabetic complications. DESIGN A cohort study based on the nationwide Diabetes in Finland database. The main analysis included 1288 women with PCOS and T2DM and 177 555 women with T2DM without PCOS (controls). Moreover, a sub-analysis that compared each woman with PCOS and T2DM to 5 controls with matching diagnosis date of T2DM and the age at onset of T2DM was conducted. METHODS The risk for diabetic complications (ie, a composite variable including retinopathy, neuropathy, nephropathy, cardiovascular, cerebrovascular or foot complications, and all-cause death) during the period 1996-2017 was analyzed using Cox regression with PCOS as the time-dependent variable and adjustment for education level, year of T2DM diagnosis, and age at T2DM diagnosis. The unadjusted cumulative incidence of diabetic complications was calculated among women with PCOS and matched controls. RESULTS The median age at T2DM diagnosis was significantly lower in women with PCOS compared with controls (33 [25th; 75th percentiles: 25; 41] versus 62 [53; 71] years). The 20-year cumulative incidence of diabetic complication after T2DM diagnosis was 35% in women with PCOS and 48% in matched controls. Women with PCOS had a smaller adjusted hazard ratio (0.70, 95% CI, 0.60-0.82) for diabetic complication. CONCLUSIONS The findings of the present register study suggest that PCOS does not seem to increase the risk of diabetic complications. However, studies with longer follow-up and clinical data, such as body mass index, are needed to verify this.
Collapse
Affiliation(s)
- Meri-Maija Ollila
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Ari Hautakoski
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Martti Arffman
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, Helsinki 00271, Finland
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Minna Koivikko
- Department of Somatics, Internal Medicine, Division of Endocrinology, Oulu University Hospital and University of Oulu, Box 23, Oulu 90029, Finland
| | - Tapani Ebeling
- Department of Somatics, Internal Medicine, Division of Endocrinology, Oulu University Hospital and University of Oulu, Box 23, Oulu 90029, Finland
| | - Reijo Sund
- Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio 70211, Finland
- Knowledge Management Unit, Kuopio University Hospital, Kuopio 70211, Finland
| | - Terhi Piltonen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| |
Collapse
|
34
|
Pesonen E, Nurkkala M, Ollila MM, Hurskainen E, Morin-Papunen LC, Jämsä T, Korpelainen R, Niemelä M, Piltonen TT. Women with polycystic ovary syndrome are at risk of emotional and uncontrolled eating at midlife: a population-based cohort study. Fertil Steril 2025; 123:383-393. [PMID: 39349119 DOI: 10.1016/j.fertnstert.2024.09.042] [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: 05/30/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024]
Abstract
OBJECTIVE To investigate eating behavior domains-emotional, uncontrolled, and cognitive restraint eating-in women with polycystic ovary syndrome (PCOS) with different PCOS phenotypes and women without PCOS at midlife. DESIGN A prospective cohort study. Eating behavior domains were assessed at age of 46 years. Predictors of eating behaviors were evaluated using variables collected at ages of 31 and 46 years. SUBJECTS Women identified as having PCOS (n = 251) at age of 31 years using the updated Rotterdam criteria were compared with women without any PCOS criteria (n = 935). The PCOS population comprised women with the classic A+B-phenotype (hyperandrogenism and oligomenorrhea, with or without elevated antimüllerian hormone, n = 60), C-phenotype (hyperandrogenism and elevated antimüllerian hormone, n = 84), and D-phenotype (oligomenorrhea and elevated antimüllerian hormone, n = 86). EXPOSURE Not applicable. MAIN OUTCOME MEASURES Revised Three-Factor Eating Questionnaire-18 scores for eating behavior domains. RESULTS Compared with women without PCOS, women with PCOS exhibited higher scores for emotional (33.1 ± 27.8 vs. 39.0 ± 29.9) and uncontrolled eating (26.7 ± 18.2 vs. 30.7 ± 19.4) but no difference in cognitive restraint (46.6 ± 18.6 vs. 45.9 ± 18.5) at age of 46 years. Emotional and uncontrolled eating scores were higher in the A+B-phenotype compared with women without PCOS, whereas uncontrolled eating scores in the C-phenotype were higher than in women without PCOS and the D-phenotype. At age of 46 years, the perception of overweight was an independent predictor of emotional eating among women with PCOS (B = 11.96 [95% confidence interval {CI}: 2.81-20.29]), whereas a history of weight loss attempts was a predictor of uncontrolled eating (B = 6.06 [95% CI: 1.05-10.83]). Among women with PCOS, higher psychological distress at age of 31 years was a significant risk factor for scoring in the highest quartile of emotional (adjusted odds ratio [aOR]: 2.85 [95% CI: 1.19-6.85]) and uncontrolled eating (aOR: 4.37 [95% CI: 1.77-10.80]) at age of 46 years. CONCLUSION Women with PCOS at midlife showed a high tendency for unfavorable eating behaviors. Our findings emphasize the need for sensitivity in weight management counseling and addressing psychological distress to prevent unfavorable eating in this population.
Collapse
Affiliation(s)
- Emilia Pesonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland; Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Marjukka Nurkkala
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Sports and Exercise Medicine, Oulu Deaconess Institute Foundation sr., Oulu, Finland; Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Meri-Maija Ollila
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Elisa Hurskainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Laure C Morin-Papunen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Timo Jämsä
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Raija Korpelainen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Sports and Exercise Medicine, Oulu Deaconess Institute Foundation sr., Oulu, Finland; Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Maisa Niemelä
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland; Centre for Wireless Communications, University of Oulu, Oulu, Finland
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| |
Collapse
|
35
|
Lee S, Aasmets O, Arffman RK, Laru J, Rossi HR, Salumets A, Piltonen TT, Org E. The reproductive tract microbiome in women with polycystic ovary syndrome and across different menstrual cycle phases. Hum Reprod 2025; 40:518-528. [PMID: 39792897 DOI: 10.1093/humrep/deae270] [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: 05/15/2024] [Revised: 10/20/2024] [Indexed: 01/12/2025] Open
Abstract
STUDY QUESTION Do polycystic ovary syndrome (PCOS), menstrual cycle phases, and ovulatory status affect reproductive tract (RT) microbiome profiles? SUMMARY ANSWER We identified microbial features associated with menstrual cycle phases in the upper and lower RT microbiome, but only two specific differences in the upper RT according to PCOS status. WHAT IS KNOWN ALREADY The vaginal and uterine microbiome profiles vary throughout the menstrual cycle. Studies have reported alterations in the vaginal microbiome among women diagnosed with PCOS. STUDY DESIGN, SIZE, DURATION This prospective case-control study included a cohort of 37 healthy control women and 52 women diagnosed with PCOS. Microbiome samples were collected from the vagina as vaginal swabs (VS) and from the uterus as endometrial flushing (EF) aspirate samples, and compared according to PCOS diagnosis, the menstrual cycle phases, and ovulatory status, at Oulu University Hospital (Oulu, Finland) from January 2017 to March 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 83 VS samples and 80 EF samples were collected. Age and body mass index (BMI) were matched between women with and without PCOS. Clinical characteristics were assessed using blood samples collected between cycle days 2 and 8, and microbial DNA was sequenced on the Ion Torrent platform. Microbial alpha diversity (i.e. the observed number of unique genera and Shannon diversity index) was analysed across sample types, PCOS diagnosis and menstrual cycle phases. Linear mixed-effects models were utilised to identify microbial features in relation to PCOS and the menstrual cycle phases. Associations between the beta diversity of the RT microbiome and PCOS- and cycle-related clinical features were calculated using PERMANOVA. MAIN RESULTS AND THE ROLE OF CHANCE Microbial alpha diversity showed no difference with PCOS (VS: Pobserved feature = 0.836, Pshannon = 0.998; EF: Pobserved feature = 0.366, Pshannon = 0.185), but varied with menstrual cycle phases (VS: Pobserved feature = 0.001, Pshannon = 0.882; EF: Pobserved feature = 0.026, Pshannon = 0.048). No difference was observed in beta diversity based on either PCOS or the menstrual cycle phases (VS: PPCOS = 0.280, Pcycle = 0.115; EF: PPCOS = 0.234, Pcycle = 0.088). In the endometrial flushing samples, we identified two novel microbial features, characterised by the ratio of differential abundance of two genera, associated with PCOS (FDR ≤ 0.1) and 13 novel features associated with the menstrual cycle phases (FDR ≤ 0.1). LIMITATIONS, REASONS FOR CAUTION Although this was the first study to simultaneously analyse, the lower and upper RT microbiome in women with and without PCOS, the limited sample size of anovulatory cases may hinder the detection of differences related to PCOS and ovulatory status. WIDER IMPLICATIONS OF THE FINDINGS The main finding suggests that PCOS and the menstrual cycle phases are associated with specific microbial features in the upper RT, indicating that the analysis of the upper RT microbiome can potentially identify biomarkers for both PCOS and menstrual cycle phases. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the Research Council of Finland (grants no. 315921, 321763, 336449), the Sigrid Jusélius Foundation, Novo Nordisk Foundation (grant no. NNF21OC0070372), and the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant (MATER, grant no. 813707). This research was also funded by the Estonian Research Council (grants no. PRG1076, PRG1414), the Horizon Europe grant (NESTOR, grant no. 101120075) of the European Commission, and EMBO Installation Grant (grant no. 3573). The funders did not participate in any processes of the study. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- S Lee
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - O Aasmets
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - R K Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - J Laru
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - H R Rossi
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - A Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| | - T T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - E Org
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| |
Collapse
|
36
|
Wang Z, Fleisch A, Rifas-Shiman SL, Calafat AM, James-Todd T, Coull BA, Chavarro JE, Hivert MF, Whooten RC, Perng W, Oken E, Mahalingaiah S. Associations of maternal per- and polyfluoroalkyl substance plasma concentrations during pregnancy with offspring polycystic ovary syndrome and related characteristics in project viva. ENVIRONMENTAL RESEARCH 2025; 268:120786. [PMID: 39798662 PMCID: PMC11839318 DOI: 10.1016/j.envres.2025.120786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) may impact ovarian folliculogenesis and steroidogenesis, but whether prenatal exposure may impact offspring reproductive health is unknown. This study examines the extent to which maternal PFAS plasma concentrations during pregnancy are associated with polycystic ovary syndrome (PCOS) and related characteristics in female offspring. METHODS We studied 322 mother-daughter pairs in Project Viva, a Boston-area longitudinal pre-birth cohort enrolled 1999-2002. We examined associations of maternal prenatal (median: 9.6 weeks gestation) plasma concentrations of six PFAS (log2 transformed) with PCOS and related characteristics among daughters during mid-to-late adolescence. We estimated the associations of single PFAS and PFAS as a mixture with each outcome, using logistic regression and quantile g-computation, respectively, adjusting for parity, and maternal sociodemographic and other lifestyle/health factors. RESULTS Among the 322 mother-daughter pairs, the majority of mothers identified as non-Hispanic White and had a college degree, and 13% of daughters had either self-reported PCOS or probable PCOS based on irregular menstrual cycles and clinical or biochemical markers of hyperandrogenism. Among all daughters, there were 27% with irregular menstrual cycles, 34% with hirsutism, and 6% with moderate-to-severe acne. When fully adjusted for confounders, per doubling of maternal 2-(N-ethyl-perfluorooctane sulfonamido) acetate (EtFOSAA) concentration was associated with higher odds of self-reported PCOS [OR (95% CI) = 2.66 (1.18, 5.99)], and per doubling of maternal perfluorononanoate (PFNA) concentration was associated with higher odds of moderate-to-severe acne [OR (95% CI) = 2.33 (1.09, 4.99)] in daughters with or without irregular menstrual cycles. We found no associations of the mixture of six PFAS with PCOS or related traits. CONCLUSION Our findings suggest a positive association between maternal concentrations of EtFOSAA and PCOS in their daughters during mid-to-late adolescence, although future studies with larger sample size and extended follow-up across the reproductive life-course are needed.
Collapse
Affiliation(s)
- Zifan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Abby Fleisch
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Westbrook, ME, USA; Pediatric Endocrinology and Diabetes, Maine Medical Center, Portland, ME, USA
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tamarra James-Todd
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brent A Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA; Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Rachel C Whooten
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Wei Perng
- Department of Epidemiology and the Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
37
|
Farhadi‐Azar M, Noroozzadeh M, Mousavi M, Saei Ghare Naz M, Ramezani Tehrani F. Impaired glucose tolerance and insulin resistance in a prenatally-androgenized rat model of polycystic ovary syndrome in later life. Exp Physiol 2025; 110:410-423. [PMID: 39613459 PMCID: PMC11868029 DOI: 10.1113/ep091912] [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: 03/25/2024] [Accepted: 10/30/2024] [Indexed: 12/01/2024]
Abstract
Polycystic ovary syndrome (PCOS), one of the most common endocrine disorders in reproductive-aged women, is associated with metabolic disturbances. The present study aimed to examine changes in body weight (BW) and glucose and insulin tolerance in a prenatally-androgenized (PNA) rat model of PCOS compared to control with increasing age. Pregnant rats in the experimental group were subcutaneously injected with 5 mg of free testosterone on the 20th day of pregnancy, while the control group received the solvent. Female offspring of both groups, PNA rats (rat model of PCOS) and control, were examined in terms of changes in BW, glucose and insulin tolerance at 3, 6, 12 and 20 months of age. BW at birth (6.53 ± 0.89 vs. 5.60 ± 1.18 g; P = 0.038), 15 (25 ± 1.15 vs. 22.36 ± 3.98 g; P = 0.019) and 30 (59.37 ± 10.19 vs.49.9 ± 9.39 g; P = 0.022) days of age was significantly increased in the rat model of PCOS compared to control, but no significant differences were observed in BW of the rat model of PCOS compared to control at 60 (P = 0.155) and 75 (P = 0.932) days or at 3 (P = 0.239), 6 (P = 0.782), 12 (P = 0.755) and 20 (P = 0.092) months of age. Rat model of PCOS showed impaired glucose tolerance (IGT) at 3 months of age (P = 0.020) and insulin resistance (IR) with increasing age (3-20 months of age) compared to control. Increased BW before puberty, IGT at 3 months of age and IR with increasing age were observed in our rat model of PCOS. This rat model may contribute to a better understanding of underlying mechanisms of changes in BW, IGT and IR in future studies.
Collapse
Affiliation(s)
- Mahbanoo Farhadi‐Azar
- Reproductive Endocrinology Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mahsa Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Maryam Mousavi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Marzieh Saei Ghare Naz
- Reproductive Endocrinology Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
- Foundation for Research & Education ExcellenceVestavia HillsALUSA
| |
Collapse
|
38
|
An J, Zhou Q, Guo X, Xu C, Jia X, Cao Z, Lu Q. From Pathophysiology to Treatment: The Role of Ferroptosis in PCOS. FRONT BIOSCI-LANDMRK 2025; 30:25586. [PMID: 40018919 DOI: 10.31083/fbl25586] [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: 07/08/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 03/01/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent gynecological endocrine and metabolic disorder in women, with an incidence rate of 10-13%. The etiology of PCOS is multifaceted, involving genetic predisposition, environmental influences, lifestyle factors, and endocrine metabolic dysregulation. Iron, a critical mineral, not only plays a role in regulating female physiological functions and the progression of PCOS but also requires careful management to avoid deficiency. However, excess iron can trigger ferroptosis, a form of nonapoptotic cell death characterized by the accumulation of lipid peroxides. While numerous studies have explored ferroptosis in patients with PCOS and animal models, the precise mechanisms and therapeutic implications remain inadequately understood. This review seeks to elucidate the pathophysiology of PCOS and the contributory factors of ferroptosis. Additionally, we examine the diverse manifestations of ferroptosis in PCOS and evaluate its role. Furthermore, we introduce ferroptosis-related traditional Chinese medicines that may enhance the understanding of PCOS pathogenesis and aid in the development of targeted therapies for ferroptosis in PCOS.
Collapse
Affiliation(s)
- Jie An
- Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Qin Zhou
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Xiaojing Guo
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Congya Xu
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - XiaoFang Jia
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Zhenzhen Cao
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Qibin Lu
- Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
- Department of Gynecology of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Ma M, Xu H, Wang K, Chen Y, Pan T, Zhu Q. Higher oxidative balance score is associated with a decreased risk of infertility: an analysis of NHANES 2013-2020. Front Nutr 2025; 12:1493253. [PMID: 39968392 PMCID: PMC11832392 DOI: 10.3389/fnut.2025.1493253] [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: 09/08/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background Oxidative stress plays a crucial role in the female reproductive system. The oxidative balance score (OBS) is a new measure for assessing the balance between antioxidant and pro-oxidative factors in diet and lifestyle. However, limited studies have explored the relationship between OBS and infertility. Methods We performed a cross-sectional study including 2,664 women aged 20-45 years, based on data from the 2013-2020 National Health and Nutrition Examination Survey (NHANES). The OBS was derived from 16 dietary components and 4 lifestyle factors. We used multivariate logistic regression analysis to examine the association between OBS and infertility. Results The analysis revealed a significant negative association between higher OBS and infertility risk, with an odds ratio (OR) of 0.98 (95% CI, 0.95-1.00) after full adjustment. Compared to the first quartile of OBS, the second, third, and fourth quartiles showed ORs of 0.71 (95% CI, 0.45-1.11), 0.79 (95% CI, 0.51-1.22), and 0.57 (95% CI, 0.35-0.92), respectively. Similarly, women in the highest dietary OBS and lifestyle OBS quartiles had a lower infertility risk compared to those in the lowest quartiles, with ORs of 0.60 (95% CI, 0.39-0.94) and 0.54 (95% CI, 0.32-0.93), respectively. Furthermore, subgroup analysis indicated that the association between the fourth quartile of OBS and infertility remained consistent, except among women with other ethnicities-including multi-racial, and college graduate or higher. Conclusion These findings suggest that high dietary OBS and lifestyle OBS are associated with a lower risk of infertility.
Collapse
Affiliation(s)
- Mingjun Ma
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Huanying Xu
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Department of TCM Gynecology, Foshan Fosun Chancheng Hospital, Foshan, Guangdong, China
| | - Kexin Wang
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yanfen Chen
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Ting Pan
- Department of TCM Gynecology, Foshan Fosun Chancheng Hospital, Foshan, Guangdong, China
| | - Qiaoling Zhu
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Department of TCM Gynecology, Foshan Fosun Chancheng Hospital, Foshan, Guangdong, China
| |
Collapse
|
40
|
Simão DO, Santos AVMP, Vieira VS, Reis FM, Cândido AL, Comim FV, Tosatti JAG, Gomes KB. Anxiety and Depression in Polycystic Ovary Syndrome: An Analysis Using the Hospital Anxiety and Depression Scale (HADS) in Women from a Low-Income Country. Exp Clin Endocrinol Diabetes 2025; 133:105-107. [PMID: 39842458 DOI: 10.1055/a-2502-8913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Affiliation(s)
- Daiane O Simão
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Vitória S Vieira
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando M Reis
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lúcia Cândido
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabio V Comim
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jéssica A G Tosatti
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karina B Gomes
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
41
|
Virtanen N, Pesonen E, Saarela U, Hurskainen E, Arffman RK, Koivunen P, Piltonen T. Association of hemoglobin levels with metabolic traits in women with PCOS. Acta Obstet Gynecol Scand 2025; 104:357-367. [PMID: 39740096 PMCID: PMC11782057 DOI: 10.1111/aogs.15047] [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: 09/02/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION Within normal variation, higher hemoglobin (Hb) levels are associated with poorer metabolic profile in population cohorts, underlying the link between oxygen delivery and cell metabolism. Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women and is commonly accompanied by metabolic derangements. In this study we sought to investigate Hb levels, and their metabolic associations, in women with PCOS. MATERIAL AND METHODS We used data from Northern Finland Birth Cohort 1966 to evaluate Hb levels in women with or without PCOS at the ages of 31 and 46 years. Linear regression models were used to investigate associations between Hb levels and essential metabolic parameters in both groups. RESULTS Women with PCOS had higher Hb levels than controls at the age of 31 years but not at the age of 46 years. Hb levels were associated positively with most of the metabolic parameters tested (body mass index, waist circumference, fasting insulin, homeostatic model assessment-insulin resistance (HOMA-IR), blood pressure, inflammatory markers, and blood lipids), with stronger associations in women with PCOS than in non-PCOS controls. There were fewer associations at the age of 46 than at 31 years, and body mass index seemed to explain many, though not all, differences between the PCOS and non-PCOS groups. CONCLUSIONS Women with PCOS have higher Hb levels at the age of 31 years. In both women with and without PCOS, Hb levels associate with poorer metabolic profile.
Collapse
Affiliation(s)
- Nikke Virtanen
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
- Faculty of Biochemistry and Molecular MedicineUniversity of OuluOuluFinland
| | - Emilia Pesonen
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
| | - Elisa Hurskainen
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
| | - Riikka K. Arffman
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
| | - Peppi Koivunen
- Faculty of Biochemistry and Molecular MedicineUniversity of OuluOuluFinland
| | - Terhi Piltonen
- Department of Obstetrics and Gynecology, Medical Research Centre, Research Unit of Clinical MedicineUniversity of Oulu, Oulu University HospitalOuluFinland
| |
Collapse
|
42
|
Dong A, Yu X, Zhang Y, Liu L, Liu F, Song W, Zheng J. Anti-Müllerian hormone regulates ovarian granulosa cell growth in PCOS rats through SMAD4. Int J Gynaecol Obstet 2025. [PMID: 39865361 DOI: 10.1002/ijgo.16184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/26/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a diverse condition with an unknown cause. The precise mechanism underlying ovulatory abnormalities in PCOS remains unclear. It is widely believed that malfunction of granulosa cells is the primary factor contributing to aberrant follicular formation in PCOS. METHODS A DHEA-induced PCOS rat model was established, and ovarian granulosa cells were extracted and identified. Anti-Müllerian hormone (AMH) and SMAD family member 4 (SMAD4) expression was detected in the serum, ovarian tissue and ovarian granulosa cells of each group, and proliferating cell nuclear antigen (PCNA), BCL2-associated 2 (BAX), cleaved caspase-3 and BCL-2 protein expression was detected by Western blot in ovarian granulosa cells. Recombinant anti-Müllerian hormone (rAMH) was administered at different concentrations to act on normal rat ovarian granulosa cells, cell proliferation was detected by cell counting kit-8 (CCK-8), apoptosis was detected by flow cytometry, and SMAD4, caspase-3, BCL-2 and cyclin A proteins were detected by Western blot. SMAD4-siRNA was transfected into rat ovarian granulosa cells of the PCOS group, and PCNA and BAX were detected by Western blot. RESULTS Compared with those in the control group, the expression of AMH and SMAD4 was increased in the ovarian tissues and granulosa cells of rats in the PCOS group, the expression of PCNA and BCL-2 proteins was decreased in the ovarian granulosa cells of the PCOS group, the expression of BAX proteins was increased, and the expression of cleaved caspase-3 was increased. Western blot results indicated that rAMH upregulated SMAD4 and caspase-3 protein expression in granulosa cells and downregulated cyclin A and BCL-2 protein expression. CCK-8 and flow cytometry results indicated that AMH decreased granulosa cells proliferation and increased apoptosis. SiRNA knockdown of SMAD4 gene increased PCNA and BCL-2 protein expression in the granulosa cells of PCOS rats and decreased BAX and cleaved caspase-3 protein expression. CONCLUSION AMH may be involved in regulating impaired ovarian granulosa cells development in PCOS rats via SMAD4.
Collapse
Affiliation(s)
- Anqi Dong
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xiaomeng Yu
- Department of Obstetrics, Women and Children's Hospital of Jinzhou, Jinzhou, Liaoning, China
| | - Yun Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lili Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Fanglin Liu
- Center for Reproductive Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wei Song
- Center for Reproductive Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jindan Zheng
- Center for Reproductive Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
43
|
Zhang Q, Yang Z, Ou X, Zhang M, Qin X, Wu G. The role of immunity in insulin resistance in patients with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2025; 15:1464561. [PMID: 39911236 PMCID: PMC11797073 DOI: 10.3389/fendo.2024.1464561] [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: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent disorder of the endocrine system with significant clinical implications, often leading to health complications related to adipose tissue accumulation, including obesity, insulin resistance (IR), metabolic syndrome, and type 2 diabetes mellitus. While the precise pathogenesis of PCOS remains unclear, it is now recognized that genetic, endocrine, and metabolic dysregulations all contribute significantly to its onset. The immunopathogenesis of PCOS has not been extensively explored, but there is growing speculation that immune system abnormalities may play a pivotal role. This chronic inflammatory state is exacerbated by factors such as obesity and hyperinsulinemia. Therefore, this review aims to elucidate the interplay between IR in PCOS patients, the controlled immune response orchestrated by immune cells and immunomodulatory molecules, and their interactions with adipocytes, hyperandrogenemia, chronic inflammation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Qixuan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhe Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Ou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengying Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyu Qin
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gengxiang Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Zhang TY, Liu NN. Fungal influence: The role of the gut mycobiome in women's health. Cell Host Microbe 2025; 33:17-19. [PMID: 39788095 DOI: 10.1016/j.chom.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025]
Abstract
In this issue of Cell Host & Microbe, Wu et al. identified enriched gut Aspergillus tubingensis in patients with polycystic ovary syndrome (PCOS). In mice, this fungus induced a PCOS-like phenotype by inhibiting interleukin (IL)-22 secretion from ILC3s via the AT-C1-AhR axis. PCOS, a women's health concern, is influenced by the gut mycobiome.
Collapse
Affiliation(s)
- Tian-Yi Zhang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
45
|
Sun J, Wang C, Zhao J, Nie X, Lu C, Ye X, He D. The effect of anterior disc displacement with polycystic ovarian syndrome on adolescent condylar bone remodeling. BMC Oral Health 2025; 25:37. [PMID: 39773212 PMCID: PMC11706174 DOI: 10.1186/s12903-024-05324-0] [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: 06/20/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Adolescent females have a high prevalence of temporomandibular joint (TMJ) anterior disc displacement (ADD), which can lead to condylar resorption and dentofacial deformity. Polycystic ovarian syndrome (PCOS) is a common endocrine disorder that disrupts bone metabolism. However, the effects of PCOS on bone remodeling especially after disc repositioning (DR) surgery are not well understood. MATERIALS AND METHODS This was a retrospective study. Patients aged 12 to 20 years diagnosed with ADD were reviewed and matched into 3 groups: A (ADD without PCOS), B (ADD with untreated PCOS), and C (ADD with treated PCOS). Each group was divided into 2 subgroups according to ADD observation (A1, B1, C1) and DR (A2, B2, C2). Condylar height (CH) was measured by MRI at the start (T0) and after more than 6 months follow-up (T1). ∆CH (T1-T0) were compared within and between groups. RESULTS 93 patients (157 joints) with an average age of 15.17 ± 2.35 years and follow-up period of 14.04 ± 9.11 months were selected in the study. ∆CH in Group B1 was significantly larger than that in Groups A1 and C1 (p = 0.048, p = 0.018). While in Group B2, it was significant smaller than Groups A2 and C2 (p < 0.001, p = 0.023). There was no significant difference of ∆CH between Groups C2 and A2. DR acquired larger ∆CH than observation within each A, B, C Groups (p < 0.05). Multiple linear regression analysis showed that ∆CH was related to the presence of PCOS (p = 0.003), PCOS treatment (p < 0.001), and DR (p < 0.001). CONCLUSIONS Adolescent ADD with untreated PCOS can aggravate condylar degeneration and affect bone remodeling after DR. PCOS treatment can improve bone remodeling.
Collapse
Affiliation(s)
- Jiali Sun
- Department of Oral Surgery, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Jiao Tong University, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chuyao Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Jiao Tong University, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jieyun Zhao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Jiao Tong University, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Xin Nie
- Biostatistics Office of Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chuan Lu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Jiao Tong University, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Xiang Ye
- Department of Gynecology and Obstetrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Dongmei He
- Department of Oral Surgery, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Jiao Tong University, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
46
|
Huang X, Geng H, Liang C, Xiong X, Du X, Zhuan Q, Liu Z, Meng L, Zhou D, Zhang L, Fu X, Qi X, Hou Y. Leonurine restrains granulosa cell ferroptosis through SLC7A11/GPX4 axis to promote the treatment of polycystic ovary syndrome. Free Radic Biol Med 2025; 226:330-347. [PMID: 39547522 DOI: 10.1016/j.freeradbiomed.2024.11.021] [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: 10/03/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder marked by ovarian dysfunction and metabolic abnormality. This study explores the therapeutic potential of leonurine (SCM-198) in PCOS. Our results show that SCM-198 treatment significantly improved ovarian function, hormone disorders and insulin resistance while reducing granulosa cell ferroptosis. This study provides the first evidence that SCM-198 modulates the gut microbiota composition, increases the abundance of Christensenella minuta, and boosts butyrate levels. Transcriptomic and metabolomic analyses revealed that PCOS patients exhibit granulosa cell ferroptosis and decreased butyrate levels in follicular fluid. Butyrate was shown to alleviate ferroptosis in granulosa cells via the SLC7A11/TXNRD1/GPX4 pathway, as confirmed in vitro with KGN cells. The therapeutic mechanism of SCM-198 in the management of PCOS via the gut microbiota-ovary axis involves the enhancement of gut microbiota and its metabolites. This intervention improves ovarian function and alleviates PCOS symptoms by targeting ferroptosis in granulosa cells.
Collapse
Affiliation(s)
- Xiaohan Huang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hucheng Geng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Chunxiao Liang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xianglei Xiong
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Xingzhu Du
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qingrui Zhuan
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhiqiang Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lin Meng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dan Zhou
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Luyao Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiangwei Fu
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xinyu Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| | - Yunpeng Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
47
|
Yang L, Chen J, Miao H, Li N, Bi H, Feng R, Miao C. The landscape of alternative splicing in granulosa cells and a potential novel role of YAP1 in PCOS. PLoS One 2024; 19:e0315750. [PMID: 39671393 PMCID: PMC11642958 DOI: 10.1371/journal.pone.0315750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent yet complex reproductive endocrine disorder affecting 11-13% of women worldwide. Its main symptoms include elevated androgen levels, irregular menstrual cycles, and long-term metabolic and offspring health implications. Despite the disease's multifaceted nature involving genetic, epigenetic, and environmental factors, the role of alternative splicing in ovarian granulosa cells remains relatively unexplored. This study aims to investigate the transcriptional and alternative splicing characteristics of granulosa cells in PCOS patients and to elucidate the potential functional consequences of these changes. Analysis of previous published transcriptome sequencing data identified 491 upregulated and 401 downregulated genes in granulosa cells of PCOS patients, significantly involved in immune-related processes. Additionally, 1250 differential splicing events, predominantly involving exon skipping and affecting 947 genes, were detected. These genes with alternative splicing patterns were found to be enriched in endoplasmic reticulum stress and protein post-translational modification processes, suggesting their role in PCOS pathology. Moreover, the study highlighted that the utilization of different splice isoforms of the YAP1 gene may impact its interaction in the Hippo signaling pathway, influencing the pathogenesis of PCOS. These findings underscore substantial alterations in alternative splicing in granulosa cells of PCOS patients, providing a novel viewpoint for comprehending the molecular underpinnings of PCOS and suggesting potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Reproductive Genetics, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Heping Hospital of Changzhi Medical College, Changzhi, China
| | - Jianhua Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Hui Miao
- Department of Reproductive Genetics, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Heping Hospital of Changzhi Medical College, Changzhi, China
| | - Na Li
- Department of Reproductive Genetics, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Heping Hospital of Changzhi Medical College, Changzhi, China
| | - Huilin Bi
- Department of Reproductive Genetics, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Heping Hospital of Changzhi Medical College, Changzhi, China
| | - Ruizhi Feng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Reproductive Medical Center of Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Innovation Center of Suzhou Nanjing Medical University, Suzhou, China
| | - Congxiu Miao
- Department of Reproductive Genetics, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Heping Hospital of Changzhi Medical College, Changzhi, China
| |
Collapse
|
48
|
Wen X, Wang L, Lv S. Follicular development and endometrial receptivity of different androgen phenotypes in women with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2024; 15:1400880. [PMID: 39726841 PMCID: PMC11669509 DOI: 10.3389/fendo.2024.1400880] [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: 03/14/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Objective Polycystic ovary syndrome (PCOS) is an important factor contributing to infertility in reproductive-aged women. Hyperandrogenism (HA) plays an important role in the pathogenesis of PCOS. This study was conducted to explore the follicular development and endometrial receptivity of different androgen phenotypes in reproductive-aged patients with PCOS. Methods A total of 268 PCOS patients with infertility were recruited and divided into two groups according to the different androgen phenotypes in this study: abnormal menstruation and hyperandrogenism (AM-HA group, n = 127) and abnormal menstruation and polycystic ovarian morphology (AM-PCOM group, n = 141). The follicular development, endometrial receptivity, pregnancy rate, and live birth rate during the natural menstrual cycle were compared between the two groups. Results The number of dominant follicles, number of ovulations, and normal ovulation rate in the AM-HA group were significantly lower compared with those in the AM-PCOM group (p < 0.05). The endometrial thickness (ET), endometrial volume (EV), vascularization index (VI), flow index (FI), and vascularization flow index (VFI) on days 14 to 24 of the menstrual cycle before ovulation were significantly lower in the AM-HA group than in the AM-PCOM group (p < 0.05). The endometrial VI, FI, and VFI, the integrin αvβ3, and VEGF concentrations in the uterine fluid during the implantation window were significantly lower in the AM-HA group compared with the AM-PCOM group (p < 0.05). However, no statistically significant differences were observed in the uterine artery blood flow parameters, ET and EV, between the two groups (p > 0.05). The biochemical pregnancy rate, clinical pregnancy rate, ongoing pregnancy rate, and live birth rate in the AM-HA group were significantly lower compared with those in the AM-PCOM group (p < 0.05). Conclusion PCOS patients with the AM-HA phenotype were vulnerable to ovulation disorders and impaired endometrial receptivity, which resulted in reduced pregnancy rate. Treatment with HA is likely to become an effective approach for improving endometrial receptivity and fecundity disorders in patients with PCOS.
Collapse
Affiliation(s)
- Xinling Wen
- Department of Anesthesiology and Operation, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Li Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shulan Lv
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
49
|
Yang M, Wang X, Zhang Y, Qian W, Tang Y. Mindfulness acting with awareness and emotional eating among polycystic ovary syndrome women with infertility: the mediating role of depression. Front Psychol 2024; 15:1499705. [PMID: 39723408 PMCID: PMC11669249 DOI: 10.3389/fpsyg.2024.1499705] [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: 09/21/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Emotional eating, characterized by the tendency to increase food intake in response to negative emotional states, is often linked to poor emotion regulation. While mindfulness-based interventions have been studied for their benefits in reducing emotional eating, less is known about how inherent mindfulness traits, relate to emotional regulation particularly among individuals with polycystic ovary syndrome (PCOS), a population known for high rates of psychological distress and disordered eating behaviors. This study investigates the associations between different facets of mindfulness, depressive symptoms, and emotional eating among individuals with PCOS and infertility who had not received any formal mindfulness intervention. A cross-sectional study was conducted involving 334 individuals. Participants completed the Five Facet Mindfulness Questionnaire-Short Form (FFMQ-SF), the Patient Health Questionnaire (PHQ-9) to assess depressive symptoms, and the Dutch Eating Behavior Questionnaire (DEBQ) to measure emotional eating. Structural Equation Modeling (SEM) was employed to examine the relationships between inherent mindfulness traits (i.e., observation, describing, acting with awareness, non-reactivity, and non-judgment), depressive symptoms, anxiety, and emotional eating. The findings indicated that the trait of acting with awareness may reduce emotional eating through its influence on depressive symptoms, while observational mindfulness was found to be associated with increased emotional eating without prior external mindfulness training. In conclusion, Mindfulness is a multidimensional construct, with its facets contributing differently to emotional regulation and eating behaviors in individuals with PCOS and infertility. Future research should explore these nuances to develop more targeted interventions.
Collapse
Affiliation(s)
- Mengye Yang
- Reproductive Center, Suzhou Municipal Hospital, Suzhou, China
- Institute of Nursing, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Wang
- Institute of Nursing, Nanjing Medical University, Nanjing, China
| | - Yan Zhang
- Reproductive Center, Suzhou Municipal Hospital, Suzhou, China
| | - Weina Qian
- Reproductive Center, Suzhou Municipal Hospital, Suzhou, China
| | - Yan Tang
- Reproductive Center, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
50
|
Chen Y, Tang YJ, Li X, Wang XM. What can we do for the adolescents with polycystic ovary syndrome? World J Pediatr 2024; 20:1205-1208. [PMID: 39614993 DOI: 10.1007/s12519-024-00857-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Yao Chen
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China
| | - Yi-Jun Tang
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China
| | - Xin Li
- Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital, School of Medicine, Fudan University, 419 Fangxie Road, Shanghai 200011, China.
| | - Xiu-Min Wang
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China.
| |
Collapse
|