1
|
Chen L, Hui L, Wang Y, Yao X, Li J. Elevated IGFBP7 expression in follicular granulosa cells promotes PCOS pathogenesis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167743. [PMID: 39988179 DOI: 10.1016/j.bbadis.2025.167743] [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: 08/03/2024] [Revised: 01/03/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
Polycystic ovary syndrome (PCOS) can result in female infertility, menstrual irregularities, metabolic disturbances, hormonal imbalances, and significantly impact the reproductive health of women of childbearing age. Hyperandrogenism and insulin resistance are typical primary endocrine features of PCOS, which are also regarded as its core pathogenesis. In this study, IGFBP7 expression in granulosa cells (GCs) from women with and without PCOS was analyzed using bulk RNA-seq. A PCOS-like mouse model was constructed using dehydroepiandrosterone in IGFBP7 knockout and wild-type mice to explore the role of IGFBP7 in PCOS. Primary GCs from mice were cultured and transfected with IGFBP7 overexpression plasmid and siRNA fragments. Proliferation, apoptosis, and steroid hormone levels were measured to investigate the effects of IGFBP7 on granulosa cells. IGFBP7 expression was found to be elevated in patients with PCOS. Following IGFBP7 knockdown in mouse GC, there was a significant increase in GC proliferation, a decrease in GC apoptosis, and a notable decrease in testosterone secretion by GC. Conversely, overexpression of IGFBP7 in mouse granulosa cells significantly inhibited GC proliferation, significantly increased GC apoptosis, and led to a marked increase in testosterone secretion by GCs. With mouse model, a reduction in PCOS symptoms in mice after IGFBP7 deletion was observed. Elevated IGFBP7 expression in PCOS granulosa cells may induce apoptosis, hinder insulin signaling, and enhance androgen synthesis. These insights offer novel avenues for understanding and treating PCOS.
Collapse
Affiliation(s)
- Li Chen
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Linhu Hui
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Yongyang Wang
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
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] [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
|
3
|
Tong C, Wu Y, Zhuang Z, Yu Y. A diagnostic model for polycystic ovary syndrome based on machine learning. Sci Rep 2025; 15:9821. [PMID: 40119083 PMCID: PMC11928512 DOI: 10.1038/s41598-025-92630-4] [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: 08/28/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Diagnosis of polycystic ovary syndrome remains a challenge. In this study, we propose constructing a diagnostic model of polycystic ovary syndrome by combining anti-Müllerian hormone with steroid hormones and oestrogens, with the aim of providing more bases and auxiliary means for the diagnosis of this disease. 1. Eighty-four samples from patients who were diagnosed with polycystic ovary syndrome at the First Affiliated Hospital of Zhejiang Chinese Medical University from May 2023 to November 2023 were collected as the case group, and 75 samples from the healthy population of the Health Screening Centre of the First Affiliated Hospital of Zhejiang Chinese Medical University during the same period were collected as the control group. 2. General information (including age, BMI, family history, medication history, etc.) and sex hormone data (including luteinising hormone, follicle stimulating hormone, prolactin, estradiol, testosterone, etc.) were collected from all study subjects. AMH and steroid hormone tests were performed on serum collected from all study subjects. 3. The data of 10 case groups and 10 control groups were randomly selected as validation set data, and the rest of the data were included in the model construction. The acquired data were screened for variables, a classification model based on a machine learning algorithm was constructed, and the constructed model was evaluated and validated for diagnostic efficacy. Ultimately, a total of 8 variables were screened and included in the subsequent model construction, namely LH, LH/FSH, E2, PRL, T, AMH, AD, and COR, with AMH having the highest diagnostic potential among all the variables included in the model. A total of five machine learning models were constructed, the logistic classification model has the best overall performance, and the support vector machine has the weakest overall performance. The validation set has an AUC of 0.86 for the model. In this study, five classification models based on machine learning algorithms were successfully constructed. Combining the evaluation metrics of each model performance, we concluded that the logistic classification model had the best performance capability in our study. However, since this study is a single-center small sample size study, some metabolic features of PCOS may be overlooked, and, as the validation set data in this study come from the same center as the modelling data, the validation results may have several limitations, so it is still necessary to expand the sample size and collect multicenter data to establish an external validation dataset to further improve the study.
Collapse
Affiliation(s)
- Cheng Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yue Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China
| | - Zhenchao Zhuang
- Adicon Clinical Laboratories, Hangzhou, 310023, Zhejiang, China.
| | - Ying Yu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
4
|
Wang J, Huang Z, Cao Z, Luo Y, Liu Y, Cao H, Tang X, Fang G. Loureirin B Reduces Insulin Resistance and Chronic Inflammation in a Rat Model of Polycystic Ovary Syndrome by Upregulating GPR120 and Activating the LKB1/AMPK Signaling Pathway. Int J Mol Sci 2024; 25:11146. [PMID: 39456928 PMCID: PMC11508921 DOI: 10.3390/ijms252011146] [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: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Polycystic ovary yndrome (PCOS) is a common metabolic disorder in women, which is usually associated with insulin resistance (IR) and chronic inflammation. Loureirin B (LrB) can effectively improve insulin resistance and alleviate chronic inflammation, and in order to investigate the therapeutic effect of LrB on polycystic ovary syndrome with insulin resistance (PCOS-IR), we conducted animal experiments. A PCOS-IR rat model was established by feeding a high-fat diet combined with letrozole (1 mg/kg·d for 21 days). The rats were treated with the GPR120 agonists TUG-891 and LrB for 4 weeks. Biochemical parameters (fasting blood glucose, total cholesterol, triglycerides, high- and low-density lipoprotein), hormone levels (serum insulin, E2, T, LH, and FSH), and inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-18) were analyzed. Histopathological analyses of ovaries were performed using hematoxylin/eosin (H&E) staining. Real-time PCR and western blotting were used to assess GPR120, NLRP3, and caspase-1 expression in ovaries, and immunohistochemistry was used to evaluate LKB1 and AMPK protein expression. LrB reduced body weight, Lee's index, ovarian index, ovarian area, and volume in PCOS-IR rats. It lowered fasting blood glucose, serum insulin, and HOMA-IR. LrB decreased total serum cholesterol, triglyceride, and LDL levels and increased HDL levels. It reduced serum T, LH, and LH/FSH and raised serum E2 and FSH levels. LrB downregulated the mRNA and protein expression levels of NLRP3 and Caspase-1, increased the protein and mRNA expression levels of GPR120 in rat ovaries, and increased LKB1 and AMPK protein expression in ovaries, ameliorating ovarian histopathological changes in PCOS-IR rats. Taken together, LrB upregulated GPR120, LKB1, and AMPK protein expression, downregulated NLRP3 and Caspase-1 protein expression, reduced insulin resistance and chronic inflammation, and ameliorated histopathological changes in ovarian tissues in PCOS rats, suggesting its potential as a treatment for PCOS.
Collapse
Affiliation(s)
- Jing Wang
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
- Health Science Center, Hubei Minzu University, Enshi 445000, China
| | - Zheng Huang
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Zhiyong Cao
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Yehao Luo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Yueting Liu
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Huilu Cao
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Xiusong Tang
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Gang Fang
- Guangxi Key Laboratory for Applied Fundamental Research of Zhuang Medicine-Key Laboratory Project under Guangxi Health Commission, Guangxi University of Chinese Medicine, Nanning 530001, China (Z.C.); (X.T.)
- Guangxi Higher Education Key Laboratory for the Research of Du-Related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China
| |
Collapse
|
5
|
Pililis S, Lampsas S, Kountouri A, Pliouta L, Korakas E, Livadas S, Thymis J, Peppa M, Kalantaridou S, Oikonomou E, Ikonomidis I, Lambadiari V. The Cardiometabolic Risk in Women with Polycystic Ovarian Syndrome (PCOS): From Pathophysiology to Diagnosis and Treatment. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1656. [PMID: 39459443 PMCID: PMC11509436 DOI: 10.3390/medicina60101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
Polycystic Ovarian Syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, with significant variations in presentation characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. Beyond reproductive health, it may also pose crucial long-term cardiometabolic risks, especially for women with specific types of PCOS, contributing to early subclinical cardiovascular atherosclerotic alterations such as endothelial dysfunction, increased arterial stiffness, and coronary artery calcium levels, respectively. Moreover, the precise relationship between clinical cardiovascular disease (CVD) and PCOS remains debated, with studies demonstrating an elevated risk while others report no significant association. This review investigates the pathophysiology of PCOS, focusing on insulin resistance and its link to subclinical and clinical cardiovascular disease. Diagnostic challenges and novel management strategies, including lifestyle interventions, medications like metformin and glucagon-like peptide-1 receptor agonists (GLP-1RAs), hormonal contraceptives, and bariatric surgery, are further discussed. Recognizing the cardiometabolic risks associated with PCOS, a comprehensive approach and early intervention should address both the reproductive and cardiometabolic dimensions of the syndrome.
Collapse
Affiliation(s)
- Sotirios Pililis
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| | - Stamatios Lampsas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
- 2nd Department of Ophthalmology, Attikon Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Aikaterini Kountouri
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| | - Loukia Pliouta
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| | - Emmanouil Korakas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| | | | - John Thymis
- 2nd Cardiology Department, Attikon University Hospital, National & Kapodistrian University of Athens, 12462 Athens, Greece; (J.T.)
| | - Melpomeni Peppa
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| | - Sophia Kalantaridou
- 3rd Department of Obstetrics and Gynecology, Attikon Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Medical School, “Sotiria” Chest Diseases Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, National & Kapodistrian University of Athens, 12462 Athens, Greece; (J.T.)
| | - Vaia Lambadiari
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (E.K.)
| |
Collapse
|
6
|
Jalil AT, Zair MA, Hanthal ZR, Naser SJ, Aslandook T, Abosaooda M, Fadhil A. Role of the AMP-Activated Protein Kinase in the Pathogenesis of Polycystic Ovary Syndrome. Indian J Clin Biochem 2024; 39:450-458. [PMID: 39346714 PMCID: PMC11436500 DOI: 10.1007/s12291-023-01139-y] [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: 04/14/2023] [Accepted: 06/09/2023] [Indexed: 10/01/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder characterized by elevated androgen levels, menstrual irregularities, and polycystic morphology of the ovaries. Affecting 6-10% of women in childbearing age, PCOS is a leading cause of infertility worldwide. In recent years, there has been a growing acknowledgment of the involvement of adenosine monophosphate-activated protein kinase (AMPK) in the development of polycystic ovary syndrome (PCOS). The expression of AMPK is diminished in polycystic ovaries, and when AMPK is silenced in human granulosa cells, there is a rise in the expression of steroidogenic enzymes, resulting in increased production of estradiol and progesterone. Additionally, in mouse models, the inhibiting AMPK intensifies the polycystic appearance of ovaries and impairs the process of ovulation. Moreover, it has been shown that AMPK activators like metformin and resveratrol ameliorate PCOS associated signs and symptoms in experimental and clinical studies. These findings, collectively, indicate the key role of AMPK in the pathogenesis of PCOS. Understanding the role of AMPK in PCOS will offer rewarding information on details of PCOS pathogenesis and will provide novel more specific therapeutic approaches. The present review summarizes the latest findings regarding the role of AMPK in PCOS obtained in experimental and clinical studies.
Collapse
Affiliation(s)
- Abduldaheem Turki Jalil
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hilla, Babylon Iraq
| | - Mahdi Abd Zair
- Department of Pharmacy, Kut University College, Kut, Wasit Iraq
| | | | - Sarmad Jaafar Naser
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Tahani Aslandook
- Department of Dentistry, Al-Turath University College, Baghdad, Iraq
| | - Munther Abosaooda
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Ali Fadhil
- Medical Laboratory Technology Department, College of Medical Technology, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
7
|
Ding H, Xiang Y, Zhu Q, Wu H, Xu T, Huang Z, Ge H. Endoplasmic reticulum stress-mediated ferroptosis in granulosa cells contributes to follicular dysfunction of polycystic ovary syndrome driven by hyperandrogenism. Reprod Biomed Online 2024; 49:104078. [PMID: 39024925 DOI: 10.1016/j.rbmo.2024.104078] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/23/2024] [Indexed: 07/20/2024]
Abstract
RESEARCH QUESTION Does hyperandrogenaemia affect the function of ovarian granulosa cells by activating ferroptosis, and could this process be regulated by endoplasmic reticulum stress? DESIGN Levels of ferroptosis and endoplasmic reticulum stress in granulosa cells were detected in women with and without polycystic ovary syndrome (PCOS) undergoing IVF. Ferroptosis and endoplasmic reticulum stress levels of ovarian tissue and follicle development were detected in control mice and PCOS-like mice models, induced by dehydroepiandrosterone. An in-vitro PCOS model of KGN cells was constructed with testosterone and ferroptosis inhibitor Fer-1. Endoplasmic reticulum stress inhibitor, tauroursodeoxycholate (TUDCA), determined the potential mechanism associated with excessive induction of ferroptosis in granulosa cells related to PCOS, and levels of ferroptosis and endoplasmic reticulum stress were detected. RESULTS Activation of ferroptosis and endoplasmic reticulum stress occurred in granulosa cells of women with PCOS and the varies of PCOS-like mice. The findings in KGN cells demonstrated that testosterone treatment results in elevation of oxidative stress levels, particularly lipid peroxidation, and intracellular iron accumulation in granulosa cells. The expression of genes and proteins associated with factors related to ferroptosis, mitochondrial membrane potential and ultrastructure showed that testosterone activated ferroptosis, whereas Fer-1 reversed these alterations. During in-vitro experiments, activation of endoplasmic reticulum stress induced by testosterone treatment was detected in granulosa cells. In granulosa cells, TUDCA, an inhibitor of endoplasmic reticulum stress, significantly mitigated testosterone-induced ferroptosis. CONCLUSIONS Ferroptosis plays a part in reproductive injury mediated by hyperandrogens associated with PCOS, and may be regulated by endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Huimin Ding
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Yu Xiang
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Qi Zhu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing Medical University, Nanjing, China
| | - Honghui Wu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Dalian Medical University, Liaoning, China
| | - Tianyue Xu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Zichao Huang
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Hongshan Ge
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China; Graduate School, Nanjing Medical University, Nanjing, China; Graduate School, Dalian Medical University, Liaoning, China.
| |
Collapse
|
8
|
Chen X, Hong L, Diao L, Yin T, Liu S. Hyperandrogenic environment regulates the function of ovarian granulosa cells by modulating macrophage polarization in PCOS. Am J Reprod Immunol 2024; 91:e13854. [PMID: 38716832 DOI: 10.1111/aji.13854] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine-metabolic disorder characterized by oligo-anovulation, hyperandrogenism, and polycystic ovaries, with hyperandrogenism being the most prominent feature of PCOS patients. However, whether excessive androgens also exist in the ovarian microenvironment of patients with PCOS, and their modulatory role on ovarian immune homeostasis and ovarian function, is not clear. METHODS Follicular fluid samples from patients participating in their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment were collected. Androgen concentration of follicular fluid was assayed by chemiluminescence, and the macrophage M1:M2 ratio was detected by flow cytometry. In an in vitro model, we examined the regulatory effects of different concentrations of androgen on macrophage differentiation and glucose metabolism levels using qRT-PCR, Simple Western and multi-factor flow cytometry assay. In a co-culture model, we assessed the effect of a hyperandrogenic environment in the presence or absence of macrophages on the function of granulosa cells using qRT-PCR, Simple Western, EdU assay, cell cycle assay, and multi-factor flow cytometry assay. RESULTS The results showed that a significantly higher androgen level and M1:M2 ratio in the follicular fluid of PCOS patients with hyperandrogenism. The hyperandrogenic environment promoted the expression of pro-inflammatory and glycolysis-related molecules and inhibited the expression of anti-inflammatory and oxidative phosphorylation-related molecules in macrophages. In the presence of macrophages, a hyperandrogenic environment significantly downregulated the function of granulosa cells. CONCLUSION There is a hyperandrogenic microenvironment in the ovary of PCOS patients with hyperandrogenism. Hyperandrogenic microenvironment can promote the activation of ovarian macrophages to M1, which may be associated with the reprogramming of macrophage glucose metabolism. The increased secretion of pro-inflammatory cytokines by macrophages in the hyperandrogenic microenvironment would impair the normal function of granulosa cells and interfere with normal ovarian follicle growth and development.
Collapse
Affiliation(s)
- Xi Chen
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Hong
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Tailang Yin
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, Wuhan, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| |
Collapse
|
9
|
Yildiz S, Moolhuijsen LME, Visser JA. The Role of Anti-Müllerian Hormone in Ovarian Function. Semin Reprod Med 2024; 42:15-24. [PMID: 38781987 DOI: 10.1055/s-0044-1786732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Anti-Müllerian hormone (AMH) is a member of the transforming growth factor β (TGFβ) superfamily, whose actions are restricted to the endocrine-reproductive system. Initially known for its role in male sex differentiation, AMH plays a role in the ovary, acting as a gatekeeper in folliculogenesis by regulating the rate of recruitment and growth of follicles. In the ovary, AMH is predominantly expressed by granulosa cells of preantral and antral follicles (i.e., post primordial follicle recruitment and prior to follicle-stimulating hormone (FSH) selection). AMH signals through a BMP-like signaling pathway in a manner distinct from other TGFβ family members. In this review, the latest insights in AMH processing, signaling, its regulation of spatial and temporal expression pattern, and functioning in folliculogenesis are summarized. In addition, effects of AMH variants on ovarian function are reviewed.
Collapse
Affiliation(s)
- Sena Yildiz
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Loes M E Moolhuijsen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Ubba V, Joseph S, Awe O, Jones D, Dsilva MK, Feng M, Wang J, Fu X, Akbar RJ, Bodnar BH, Hu W, Wang H, Yang X, Yang L, Yang P, Taib B, Ahima R, Divall S, Wu S. Reproductive Profile of Neuronal Androgen Receptor Knockout Female Mice With a Low Dose of DHT. Endocrinology 2024; 165:bqad199. [PMID: 38156784 PMCID: PMC10794876 DOI: 10.1210/endocr/bqad199] [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: 05/05/2023] [Revised: 09/08/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Hyperandrogenism and polycystic ovarian syndrome result from the imbalance or increase of androgen levels in females. Androgen receptor (AR) mediates the effects of androgens, and this study examines whether neuronal AR plays a role in reproduction under normal and increased androgen conditions in female mice. The neuron-specific AR knockout (KO) mouse (SynARKO) was generated from a female mouse (synapsin promoter driven Cre) and a male mouse (Ar fl/y). Puberty onset and the levels of reproductive hormones such as LH, FSH, testosterone, and estradiol were comparable between the control and the SynARKO mice. There were no differences in cyclicity and fertility between the control and SynARKO mice, with similar impairment in both groups on DHT treatment. Neuronal AR KO, as in this SynARKO mouse model, did not alleviate the infertility associated with DHT treatment. These studies suggest that neuronal AR KO neither altered reproductive function under physiological androgen levels, nor restored fertility under hyperandrogenic conditions.
Collapse
Affiliation(s)
- Vaibhave Ubba
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Serene Joseph
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Dustin Jones
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Milan K Dsilva
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Mingxiao Feng
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
| | - Junjiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
- Departments of Gastrointestinal Surgery and General Surgery, Guangdong Provincial People’s Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Xiaomin Fu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Razeen J Akbar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Brittany H Bodnar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Wenhui Hu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Ling Yang
- Department of Medical Genetics & Molecular Biochemistry, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, USA
| | - Bouchra Taib
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Rexford Ahima
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle’s Children’s Hospital, Seattle, WA, 98145-5005, USA
| | - Sheng Wu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
| |
Collapse
|
11
|
Khatun M, Lundin K, Naillat F, Loog L, Saarela U, Tuuri T, Salumets A, Piltonen TT, Tapanainen JS. Induced Pluripotent Stem Cells as a Possible Approach for Exploring the Pathophysiology of Polycystic Ovary Syndrome (PCOS). Stem Cell Rev Rep 2024; 20:67-87. [PMID: 37768523 PMCID: PMC10799779 DOI: 10.1007/s12015-023-10627-w] [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] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine condition among women with pleiotropic sequelae possessing reproductive, metabolic, and psychological characteristics. Although the exact origin of PCOS is elusive, it is known to be a complex multigenic disorder with a genetic, epigenetic, and environmental background. However, the pathogenesis of PCOS, and the role of genetic variants in increasing the risk of the condition, are still unknown due to the lack of an appropriate study model. Since the debut of induced pluripotent stem cell (iPSC) technology, the ability of reprogrammed somatic cells to self-renew and their potential for multidirectional differentiation have made them excellent tools to study different disease mechanisms. Recently, researchers have succeeded in establishing human in vitro PCOS disease models utilizing iPSC lines from heterogeneous PCOS patient groups (iPSCPCOS). The current review sets out to summarize, for the first time, our current knowledge of the implications and challenges of iPSC technology in comprehending PCOS pathogenesis and tissue-specific disease mechanisms. Additionally, we suggest that the analysis of polygenic risk prediction based on genome-wide association studies (GWAS) could, theoretically, be utilized when creating iPSC lines as an additional research tool to identify women who are genetically susceptible to PCOS. Taken together, iPSCPCOS may provide a new paradigm for the exploration of PCOS tissue-specific disease mechanisms.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland.
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Florence Naillat
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Liisa Loog
- Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, 50406, Estonia
- Competence Centre of Health Technologies, Tartu, 50411, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, 14186, Sweden
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
- Department of Obstetrics and Gynecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
12
|
Marie C, Pierre A, Mayeur A, Giton F, Corre R, Grynberg M, Cohen-Tannoudji J, Guigon CJ, Chauvin S. Dysfunction of Human Estrogen Signaling as a Novel Molecular Signature of Polycystic Ovary Syndrome. Int J Mol Sci 2023; 24:16689. [PMID: 38069013 PMCID: PMC10706349 DOI: 10.3390/ijms242316689] [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] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Estradiol (E2) is a major hormone-controlling folliculogenesis whose dysfunction may participate in polycystic ovary syndrome (PCOS) infertility. To determine whether both the concentration and action of E2 could be impaired in non-hyperandrogenic overweight PCOS women, we isolated granulosa cells (GCs) and follicular fluid (FF) from follicles of women undergoing ovarian stimulation (27 with PCOS, and 54 without PCOS). An analysis of the transcript abundance of 16 genes in GCs showed that androgen and progesterone receptor expressions were significantly increased in GCs of PCOS (by 2.7-fold and 1.5-fold, respectively), while those of the steroidogenic enzymes CYP11A1 and HSD3B2 were down-regulated (by 56% and 38%, respectively). Remarkably, treatment of GC cultures with E2 revealed its ineffectiveness in regulating the expression of several key endocrine genes (e.g., GREB1 or BCL2) in PCOS. Additionally, a comparison of the steroid concentrations (measured by GC/MS) in GCs with those in FF of matched follicles demonstrated that the significant decline in the E2 concentration (by 23%) in PCOS FF was not the result of the E2 biosynthesis reduction. Overall, our study provides novel hallmarks of PCOS by highlighting the ineffective E2 signaling in GCs as well as the dysregulation in the expression of genes involved in follicular growth, which may contribute to aberrant folliculogenesis in non-hyperandrogenic women with PCOS.
Collapse
Affiliation(s)
- Clémentine Marie
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| | - Alice Pierre
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| | - Anne Mayeur
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Antoine Béclère, 92140 Clamart, France;
| | - Frank Giton
- AP-HP, Pôle Biologie-Pathologie Henri Mondor, Inserm IMRB U955, 94010 Créteil, France;
| | - Raphael Corre
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| | - Michaël Grynberg
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Antoine Béclère, 92140 Clamart, France;
| | - Joëlle Cohen-Tannoudji
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| | - Céline J. Guigon
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| | - Stéphanie Chauvin
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; (C.M.); (A.P.); (R.C.); (M.G.); (J.C.-T.); (C.J.G.)
| |
Collapse
|
13
|
Wang X, Zhu R, Han H, Jin J. Body Fat Distribution and Female Infertility: a Cross-Sectional Analysis Among US Women. Reprod Sci 2023; 30:3243-3252. [PMID: 37277689 DOI: 10.1007/s43032-023-01280-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
Abstract
At present, the effect of body fat distribution on female reproductive health is still inconclusive. The purpose of our study was to analyze the correlation between female infertility rates and the fat mass portion of the android region to the gynoid region (the A/G ratio) among US women of reproductive age. Female infertility is defined as a failure to get pregnant after 12 months of unprotected sexual activity. A total of 3434 women of reproductive age were included in this study as part of the 2013-2018 National Health and Nutrition Examination Survey (NHANES). The A/G ratio was used to assess the body fat distribution of participants. Based on the comprehensive study design and sample weights, it was determined that the A/G ratio was associated with female infertility primarily through logistic regression analyses. After adjusting for potential confounders, the multivariate regression analysis indicated an increase in the A/G ratio was correlated with an increase in the prevalence of female infertility (OR = 4.374, 95% CI:1.809-10.575). Subgroup analyses showed an increased prevalence of infertility in non-Hispanic Whites (P = 0.012), non-diabetic individuals (P = 0.008), individuals under 35 years old (P = 0.002), and individuals with secondary infertility (P = 0.01). The trend tests and smooth curve fitting illustrate a linear trend between the A/G ratio and female infertility. Future researches are warranted to confirm the causal relationship between body fat distribution and female infertility, which may provide an insight into future prevention and treatment of female infertility.
Collapse
Affiliation(s)
- Xinzhe Wang
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Qinhuai District, Nanjing, 210000, Jiangsu Province, People's Republic of China
| | - Rui Zhu
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Qinhuai District, Nanjing, 210000, Jiangsu Province, People's Republic of China
| | - Huawei Han
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Jin
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Qinhuai District, Nanjing, 210000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
14
|
Fahs D, Salloum D, Nasrallah M, Ghazeeri G. Polycystic Ovary Syndrome: Pathophysiology and Controversies in Diagnosis. Diagnostics (Basel) 2023; 13:diagnostics13091559. [PMID: 37174950 PMCID: PMC10177792 DOI: 10.3390/diagnostics13091559] [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: 02/16/2023] [Revised: 03/16/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex and heterogeneous disorder that commonly affects women in the reproductive age group. The disorder has features that propose a blend of functional reproductive disorders, such as anovulation and hyperandrogenism, and metabolic disorders, such as hyperglycemia, hypertension, and obesity in women. Until today, the three implemented groups of criteria for the diagnosis of PCOS are from the National Institutes of Health (NIH) in the 1990s, Rotterdam 2003, and the Androgen Excess Polycystic Ovary Syndrome 2009 criteria. Currently, the most widely utilized criteria are the 2003 Rotterdam criteria, which validate the diagnosis of PCOS with the incidence of two out of the three criteria: hyperandrogenism (clinical and/or biochemical), irregular cycles, and polycystic ovary morphology. Currently, the anti-Müllerian hormone in serum is introduced as a substitute for the follicular count and is controversially emerging as an official polycystic ovarian morphology/PCOS marker. In adolescents, the two crucial factors for PCOS diagnosis are hyperandrogenism and irregular cycles. Recently, artificial intelligence, specifically machine learning, is being introduced as a promising diagnostic and predictive tool for PCOS with minimal to zero error that would help in clinical decisions regarding early management and treatment. Throughout this review, we focused on the pathophysiology, clinical features, and diagnostic challenges in females with PCOS.
Collapse
Affiliation(s)
- Duaa Fahs
- Department of Obstetrics and Gynecology, Faculty of Medicine, American University of Beirut Medical Center, Beirut P.O. Box 113-6044, Lebanon
| | - Dima Salloum
- Department of Obstetrics and Gynecology, Faculty of Medicine, American University of Beirut Medical Center, Beirut P.O. Box 113-6044, Lebanon
| | - Mona Nasrallah
- Division of Endocrinology and Metabolism, Faculty of Medicine, American University of Beirut Medical Center, Beirut P.O. Box 113-6044, Lebanon
| | - Ghina Ghazeeri
- Department of Obstetrics and Gynecology, Faculty of Medicine, American University of Beirut Medical Center, Beirut P.O. Box 113-6044, Lebanon
| |
Collapse
|
15
|
Zeng X, Zhong Q, Li M, Liu Y, long S, Xie Y, Mo Z. Androgen increases klotho expression via the androgen receptor-mediated pathway to induce GCs apoptosis. J Ovarian Res 2023; 16:10. [PMID: 36641458 PMCID: PMC9840339 DOI: 10.1186/s13048-022-01087-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/20/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Many epidemiological studies have shown that anovulatory polycystic ovary syndrome (PCOS) is accompanied by hyperandrogenism. However, the exact mechanism of hyperandrogen-induced anovulation remains to be elucidated. In this study, we aimed to investigate the potential mechanism of anovulation in PCOS. To investigate the role of klotho as a key factor in the androgen receptor (AR)-mediated development of PCOS, we investigated the effects of testosterone on ovarian klotho expression in vivo and in vitro. RESULTS Testosterone propionate (TP)-induced rats showed cycle irregularity, hyperandrogenism, polycystic ovarian changes, dyslipidemia. However, inhibition of AR expression could relieve PCOS traits. We also found that AR and klotho showed relatively high expression in PCOS rat ovarian tissue and in TP-induced granulosa cells (GCs), which was inhibited by the addition of flutamide. TP-induced GCs apoptosis was suppressed by AR antagonist, as well as silencing klotho expression in human GCs. Chromatin immunoprecipitation assay demonstrated that AR indirectly binds to the klotho promoter. CONCLUSIONS Our results demonstrated TP mediates the expression of klotho via androgen receptor and klotho alterations could be a reason for ovarian dysfunction in PCOS.
Collapse
Affiliation(s)
- Xin Zeng
- grid.443385.d0000 0004 1798 9548Guangxi Key Laboratory of Diabetic Systems Medicine, Guangxi Province Postgraduate Co-Training Base for Cooperative Innovation in Basic Medicine, Guilin Medical University, Guilin, 541199 Guangxi China ,Department of Basic Medicine, Chongqing College of Traditional Chinese Medicine, Chongqing, 402760 People’s Republic of China
| | - Qiaoqing Zhong
- grid.452223.00000 0004 1757 7615Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, 410008 China ,grid.38142.3c000000041936754XDepartment of Anesthesia, Critical Care & Pain Med, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Ming Li
- grid.443385.d0000 0004 1798 9548Guangxi Key Laboratory of Diabetic Systems Medicine, Guangxi Province Postgraduate Co-Training Base for Cooperative Innovation in Basic Medicine, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Yating Liu
- grid.412017.10000 0001 0266 8918Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001 China
| | - Shuanglian long
- grid.412017.10000 0001 0266 8918Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001 China
| | - Yuanjie Xie
- grid.443385.d0000 0004 1798 9548Guangxi Key Laboratory of Diabetic Systems Medicine, Guangxi Province Postgraduate Co-Training Base for Cooperative Innovation in Basic Medicine, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Zhongcheng Mo
- grid.443385.d0000 0004 1798 9548Guangxi Key Laboratory of Diabetic Systems Medicine, Guangxi Province Postgraduate Co-Training Base for Cooperative Innovation in Basic Medicine, Guilin Medical University, Guilin, 541199 Guangxi China
| |
Collapse
|
16
|
Khamoshina MB, Artemenko YS, Bayramova AA, Ryabova VA, Orazov MR. Polycystic ovary syndrome and obesity: a modern paradigm. RUDN JOURNAL OF MEDICINE 2022. [DOI: 10.22363/2313-0245-2022-26-4-382-395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Polycystic ovary syndrome is a heterogeneous endocrine disease that affects women of childbearing age. The pathogenesis of polycystic ovary syndrome has not been fully studied to date, its paradigm considers the genetic determinism of the manifestation of hormonal and metabolic disorders, which are considered to be criteria for the verification of the disease (hyperandrogenism, oligo/anovulation and/or polycystic ovarian transformation during ultrasound examination (ultrasound). This review discusses the main ways of interaction between hyperandrogenism, insulin resistance and obesity and their role in the pathogenesis of polycystic ovary syndrome, as well as possible methods of treatment for this category of patients. The review analyzes the role of hyperandrogenism and insulin resistance in the implementation of the genetic scenario of polycystic ovary syndrome and finds out the reasons why women with polycystic ovary syndrome often demonstrate the presence of a «metabolic trio» - hyperinsulinemia, insulin resistance and type 2 diabetes mellitus. It is noted that obesity is not included in the criteria for the diagnosis of polycystic ovary syndrome, but epidemiological data confirm the existence of a relationship between these diseases. Obesity, especially visceral, which is often found in women with polycystic ovary syndrome, enhances and worsens metabolic and reproductive outcomes with polycystic ovary syndrome, as well as increases insulin resistance and compensatory hyperinsulinemia, which, in turn, stimulates adipogenesis and suppresses lipolysis. Obesity increases the sensitivity of tech cells to luteinizing hormone stimulation and enhances functional hyperandrogenism of the ovaries, increasing the production of androgens by the ovaries. Excess body weight is associated with a large number of inflammatory adipokines, which, in turn, contribute to the growth of insulin resistance and adipogenesis. Obesity and insulin resistance exacerbate the symptoms of hyperandrogenism, forming a vicious circle that contributes to the development of polycystic ovary syndrome. These data allow us to conclude that bariatric surgery can become an alternative to drugs (metformin, thiazolidinedione analogs of glucagon-like peptide-1), which has shown positive results in the treatment of patients with polycystic ovary syndrome and obesity.
Collapse
|
17
|
Aydogan Mathyk B, Cetin E, Yildiz BO. Use of anti-Müllerian hormone for understanding ovulatory dysfunction in polycystic ovarian syndrome. Curr Opin Endocrinol Diabetes Obes 2022; 29:528-534. [PMID: 36218229 DOI: 10.1097/med.0000000000000772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to understand how anti-Müllerian hormone (AMH) contributes to ovulatory dysfunction in polycystic ovarian syndrome (PCOS). RECENT FINDINGS In the last few years, new findings have emerged on AMH and its role on the central nervous system causing ovulatory dysfunction. SUMMARY Anovulation is a prominent feature of PCOS. Women with anovulatory PCOS have higher AMH levels than in ovulatory PCOS. Higher levels of AMH may contribute to the pathophysiology of PCOS through central and peripheral actions. Once universal standardization is achieved to measure serum AMH, the benefits would be significant in diagnosing women with PCOS.
Collapse
Affiliation(s)
- Begum Aydogan Mathyk
- Department of Obstetrics and Gynecology, HCA/University of South Florida Morsani College of Medicine GME, Brandon Regional Hospital, Brandon, Florida
| | - Esra Cetin
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois, USA
| | - Bulent O Yildiz
- Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Hacettepe, Ankara, Turkey
| |
Collapse
|
18
|
Esencan E, Beroukhim G, Seifer DB. Age-related changes in Folliculogenesis and potential modifiers to improve fertility outcomes - A narrative review. Reprod Biol Endocrinol 2022; 20:156. [PMID: 36397149 PMCID: PMC9670479 DOI: 10.1186/s12958-022-01033-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022] Open
Abstract
Reproductive aging is characterized by a decline in oocyte quantity and quality, which is directly associated with a decline in reproductive potential, as well as poorer reproductive success and obstetrical outcomes. As women delay childbearing, understanding the mechanisms of ovarian aging and follicular depletion have become increasingly more relevant. Age-related meiotic errors in oocytes are well established. In addition, it is also important to understand how intraovarian regulators change with aging and how certain treatments can mitigate the impact of aging. Individual studies have demonstrated that reproductive pathways involving antimullerian hormone (AMH), vascular endothelial growth factor (VEGF), neurotropins, insulin-like growth factor 1 (IGF1), and mitochondrial function are pivotal for healthy oocyte and cumulus cell development and are altered with increasing age. We provide a comprehensive review of these individual studies and explain how these factors change in oocytes, cumulus cells, and follicular fluid. We also summarize how modifiers of folliculogenesis, such as vitamin D, coenzyme Q, and dehydroepiandrosterone (DHEA) may be used to potentially overcome age-related changes and enhance fertility outcomes of aged follicles, as evidenced by human and rodent studies.
Collapse
Affiliation(s)
- Ecem Esencan
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA.
| | - Gabriela Beroukhim
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| | - David B Seifer
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| |
Collapse
|
19
|
Walters KA, Moreno-Asso A, Stepto NK, Pankhurst MW, Rodriguez Paris V, Rodgers RJ. Key signalling pathways underlying the aetiology of polycystic ovary syndrome. J Endocrinol 2022; 255:R1-R26. [PMID: 35980384 DOI: 10.1530/joe-22-0059] [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: 06/09/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine condition characterised by a range of reproductive, endocrine, metabolic and psychological abnormalities. Reports estimate that around 10% of women of reproductive age are affected by PCOS, representing a significant prevalence worldwide, which poses a high economic health burden. As the origin of PCOS remains largely unknown, there is neither a cure nor mechanism-based treatments leaving patient management suboptimal and focused solely on symptomatic treatment. However, if the underlying mechanisms underpinning the development of PCOS were uncovered then this would pave the way for the development of new interventions for PCOS. Recently, there have been significant advances in our understanding of the underlying pathways likely involved in PCOS pathogenesis. Key insights include the potential involvement of androgens, insulin, anti-Müllerian hormone and transforming growth factor beta in the development of PCOS. This review will summarise the significant scientific discoveries on these factors that have enhanced our knowledge of the mechanisms involved in the development of PCOS and discuss the impact these insights may have in shaping the future development of effective strategies for women with PCOS.
Collapse
Affiliation(s)
- Kirsty A Walters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Alba Moreno-Asso
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
| | - Nigel K Stepto
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
- Monash Centre for Health Research and Implementation, Monash University and Monash Health, Clayton, Victoria, Australia
- Medicine at Western Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Valentina Rodriguez Paris
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Raymond J Rodgers
- The Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
20
|
Association of Insulin Resistance and Elevated Androgen Levels with Polycystic Ovarian Syndrome (PCOS): A Review of Literature. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9240569. [PMID: 35356614 PMCID: PMC8959968 DOI: 10.1155/2022/9240569] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/14/2022]
Abstract
The polycystic ovary syndrome (PCOS) is the disease featured by elevated levels of androgens, ovulatory dysfunction, and morphological abnormalities. At reproductive stage of women, the rate of PCOS occurrence is measured as 6–10% and the prevalence rate may be double. There are different pathophysiological factors involved in PCOS, and they play a major role in various abnormalities in individual patient. It is clear that there is noteworthy elevation of androgen in PCOS, causing substantial misery and infertility problems. The overexposure of androgen is directly linked with insulin resistance and hyperinsulinaemia. It has been reported previously that PCOS is related to cardiac metabolic miseries and potently increases the risk of heart diseases. Endometrial cancer is also a serious concern which is reported with exceedingly high incidence in women with PCOS. However, the overexposure of androgen has direct and specific influence on the development of insulin resistance. Although many factors are involved, resistance to the insulin and enhanced level of androgen are considered the major causes of PCOS. In the present review, we have focused on the pathophysiology and major revolutions of insulin resistance and excessive levels of androgen in females with PCOS.
Collapse
|
21
|
Anti-Müllerian hormone has limited ability to predict fecundability in Chinese women: a preconception cohort study. Reprod Biomed Online 2022; 44:1055-1063. [DOI: 10.1016/j.rbmo.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/09/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
|
22
|
Chauvin S, Cohen-Tannoudji J, Guigon CJ. Estradiol Signaling at the Heart of Folliculogenesis: Its Potential Deregulation in Human Ovarian Pathologies. Int J Mol Sci 2022; 23:ijms23010512. [PMID: 35008938 PMCID: PMC8745567 DOI: 10.3390/ijms23010512] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/26/2023] Open
Abstract
Estradiol (E2) is a major hormone controlling women fertility, in particular folliculogenesis. This steroid, which is locally produced by granulosa cells (GC) within ovarian follicles, controls the development and selection of dominant preovulatory follicles. E2 effects rely on a complex set of nuclear and extra-nuclear signal transduction pathways principally triggered by its nuclear receptors, ERα and ERβ. These transcription factors are differentially expressed within follicles, with ERβ being the predominant ER in GC. Several ERβ splice isoforms have been identified and display specific structural features, which greatly complicates the nature of ERβ-mediated E2 signaling. This review aims at providing a concise overview of the main actions of E2 during follicular growth, maturation, and selection in human. It also describes the current understanding of the various roles of ERβ splice isoforms, especially their influence on cell fate. We finally discuss how E2 signaling deregulation could participate in two ovarian pathogeneses characterized by either a follicular arrest, as in polycystic ovary syndrome, or an excess of GC survival and proliferation, leading to granulosa cell tumors. This review emphasizes the need for further research to better understand the molecular basis of E2 signaling throughout folliculogenesis and to improve the efficiency of ovarian-related disease therapies.
Collapse
|
23
|
Moolhuijsen LME, Louwers YV, McLuskey A, Broer L, Uitterlinden AG, Verdiesen RMG, Sisk RK, Dunaif A, Laven JSE, Visser JA. OUP accepted manuscript. Hum Reprod 2022; 37:1544-1556. [PMID: 35451015 PMCID: PMC9247424 DOI: 10.1093/humrep/deac082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 03/29/2022] [Indexed: 11/19/2022] Open
Abstract
STUDY QUESTION Do polymorphisms in the anti-Müllerian hormone (AMH) promoter have an effect on AMH levels in patients with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER We have identified a novel AMH promoter polymorphism rs10406324 that is associated with lower serum AMH levels and is suggested to play a role in the mechanism of regulation of AMH gene expression in women. WHAT IS KNOWN ALREADY Follicle number is positively correlated with serum AMH levels, reflected by elevated AMH levels in women with PCOS. In addition, it is suggested that AMH production per follicle is higher in women with PCOS than in normo-ovulatory women, implying an altered regulation of AMH in PCOS. STUDY DESIGN, SIZE, DURATION A discovery cohort of 655 PCOS women of Northern European ancestry and both an internal and external validation PCOS cohort (n = 458 and n = 321, respectively) were included in this study. Summary-level data of an AMH genome-wide association study meta-analysis including 7049 normo-ovulatory women was included as a control cohort. A genetic approach was taken through association analysis and in silico analysis of the associated variants in the AMH promoter. In vitro analysis was performed to investigate the functional mechanisms. PARTICIPANTS/MATERIALS, SETTING, METHODS All common two-allelic single-nucleotide polymorphisms (SNPs) in the region Chr19:2 245 353–2 250 827 bp (Build 37) were selected for the analysis. Linear regression analyses were performed to determine the association between SNPs in the AMH promoter region and serum AMH levels. For the in silico analysis, the webtools ‘HaploReg’ v4.1 for ENCODE prediction weight matrices and ‘atSNP’ were used. In vitro analysis was performed using KK1 cells, a mouse granulosa cell line and COV434 cells, a human granulosa tumor cell line. Cells were transfected with the reference or the variant human AMH promoter reporter construct together with several transcription factors (TFs). Dual-Glo® Luciferase Assay was performed to measure the luciferase activity. MAIN RESULTS AND THE ROLE OF CHANCE Polymorphism rs10406324 was significantly associated with serum AMH levels in all three PCOS cohorts. Carriers of the minor allele G had significantly lower log-transformed serum AMH levels compared to non-carriers (P = 8.58 × 10−8, P = 1.35 × 10−3 and P = 1.24 × 10−3, respectively). This result was validated in a subsequent meta-analysis (P = 3.24 × 10−12). Interestingly, rs10406324 was not associated with follicle count, nor with other clinical traits. Also, in normo-ovulatory women, the minor allele of this variant was associated with lower serum AMH levels (P = 1.04 × 10−5). These findings suggest that polymorphism rs10406324 plays a role in the regulation of AMH expression, irrespective of clinical background. In silico analysis suggested a decreased binding affinity of the TFs steroidogenenic factor 1, estrogen-related receptor alpha and glucocorticoid receptor to the minor allele G variant, however in vitro analysis did not show a difference in promoter activity between the A and G allele. LIMITATIONS, REASONS FOR CAUTION Functional analyses were performed in a mouse and a human granulosa cell line using an AMH promoter reporter construct. This may have limited assessment of the impact of the polymorphism on higher order chromatin structures. Human granulosa cells generated from induced pluripotent stem cells, combined with gene editing, may provide a method to elucidate the exact mechanism behind the decrease in serum AMH levels in carriers of the −210 G allele. We acknowledge that the lack of follicle number in the external validation and the control cohort is a limitation of the paper. Although we observed that the association between rs10406324 and AMH levels was independent of follicle number in our discovery and internal validation PCOS cohorts, we cannot fully rule out that the observed effects on serum AMH levels are, in part, caused by differences in follicle number. WIDER IMPLICATIONS OF THE FINDINGS These results suggest that variations in serum AMH levels are not only caused by differences in follicle number but also by genetic factors. Therefore, the genetic context should be taken into consideration when assessing serum AMH levels in women. This may have clinical consequences when serum AMH levels are used as a marker for the polycystic ovarian morphology phenotype. STUDY FUNDING/COMPETING INTEREST(S) No external funding was used. J.S.E.L. has received consultancy fees from the following companies: Ferring, Roche Diagnostics and Ansh Labs and has received travel reimbursement from Ferring. J.A.V. has received royalties from AMH assays, paid to the institute/lab with no personal financial gain. The other authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Loes M E Moolhuijsen
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Yvonne V Louwers
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Anke McLuskey
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Renée M G Verdiesen
- Division of Molecular Pathology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ryan K Sisk
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joop S E Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
24
|
Chen W, Pang Y. Metabolic Syndrome and PCOS: Pathogenesis and the Role of Metabolites. Metabolites 2021; 11:metabo11120869. [PMID: 34940628 PMCID: PMC8709086 DOI: 10.3390/metabo11120869] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases among women of reproductive age and is associated with many metabolic manifestations, such as obesity, insulin resistance (IR) and hyperandrogenism. The underlying pathogenesis of these metabolic symptoms has not yet been fully elucidated. With the application of metabolomics techniques, a variety of metabolite changes have been observed in the serum and follicular fluid (FF) of PCOS patients and animal models. Changes in metabolites result from the daily diet and occur during uncommon physiological routines. However, some of these metabolite changes may provide evidence to explain possible mechanisms and new approaches for prevention and therapy. This article reviews the pathogenesis of PCOS metabolic symptoms and the relationship between metabolites and the pathophysiology of PCOS. Furthermore, the potential clinical application of some specific metabolites will be discussed.
Collapse
Affiliation(s)
- Weixuan Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Yanli Pang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
- Correspondence:
| |
Collapse
|
25
|
Cluzet V, Devillers MM, Petit F, Pierre A, Giton F, Airaud E, L'Hôte D, Leary A, Genestie C, Treilleux I, Mayeur A, Katzenellenbogen JA, Kim SH, Cohen-Tannoudji J, Chauvin S, Guigon CJ. Estradiol promotes cell survival and induces Greb1 expression in granulosa cell tumors of the ovary through an ERα-dependent mechanism. J Pathol 2021; 256:335-348. [PMID: 34860414 DOI: 10.1002/path.5843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/10/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
Abstract
Granulosa cell tumor (GCT) is a form of ovarian tumor characterized by its tendency to recur years after surgical ablation. Little is known on the mechanisms involved in GCT development and progression. GCTs can produce estradiol (E2), but whether this hormone could play a role in this cancer through its nuclear receptors, i.e., ERα and ERβ, remains unknown. Here, we addressed this issue by cell-based and molecular studies on human GCTs and GCT cell lines. Importantly, we observed that E2 significantly increased the growth of GCT cells by promoting cell survival. The use of selective agonists of each type of receptor, together with Esr1 (ERα) or Esr2 (ERβ)-deleted GCT cells revealed that E2 mediated its effects through ERα-dependent genomic mechanisms and ERβ/ERα-dependent extra-nuclear mechanisms. Notably, the expression of Greb1, a prototypical ER target gene, was dose-dependently up-regulated by E2 specifically through ERα in GCT cells. Accordingly, using GCTs from patients, we found that GREB1 mRNA abundance was positively correlated to intra-tumoral E2 concentrations. Tissue microarrays analyses showed that there were various combinations of ER expression in primary and recurrent GCTs, and that ERα expression persisted only in combination with ERβ in ~40% of recurrent tumors. Altogether, this study demonstrates that E2 can promote the progression of GCTs, with a clear dependence on ERα. In addition to demonstrating that GCTs can be classified as a hormone-related cancer, our results also highlight that the nature of ER forms present in recurrent GCTs could underlie the variable efficiency of endocrine therapies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Victoria Cluzet
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Marie M Devillers
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Florence Petit
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Alice Pierre
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Frank Giton
- AP-HP, Pôle biologie-Pathologie Henri Mondor, INSERM IMRB U955, Créteil, France
| | - Eloïse Airaud
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - David L'Hôte
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Alexandra Leary
- Gustave Roussy Cancer Campus and University of Paris-Saclay, Villejuif, France
| | - Catherine Genestie
- Department of Pathology, University Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Anne Mayeur
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Antoine Béclère, Clamart, France
| | - John A Katzenellenbogen
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Sung Hoon Kim
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | | | - Stéphanie Chauvin
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Céline J Guigon
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| |
Collapse
|
26
|
Anti-Müllerian Hormone in Pathogenesis, Diagnostic and Treatment of PCOS. Int J Mol Sci 2021; 22:ijms222212507. [PMID: 34830389 PMCID: PMC8619458 DOI: 10.3390/ijms222212507] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/19/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-aged women. It is characterized by chronic anovulation, hyperandrogenism, and the presence of polycystic ovary in ultrasound examination. PCOS is specified by an increased number of follicles at all growing stages, mainly seen in the preantral and small antral follicles and an increased serum level of Anti-Müllerian Hormone (AMH). Because of the strong correlation between circulating AMH levels and antral follicle count on ultrasound, Anti-Müllerian Hormone has been proposed as an alternative marker of ovulatory dysfunction in PCOS. However, the results from the current literature are not homogeneous, and the specific threshold of AMH in PCOS and PCOM is, therefore, very challenging. This review aims to update the current knowledge about AMH, the pathophysiology of AMH in the pathogenesis of PCOS, and the role of Anti-Müllerian Hormone in the treatment of this syndrome.
Collapse
|
27
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
28
|
Wu LM, Wang YX, Zhan Y, Liu AH, Wang YX, Shen HF, Wang YF, Wang LY, Tao ZB, Wang YQ. Dulaglutide, a long-acting GLP-1 receptor agonist, can improve hyperandrogenemia and ovarian function in DHEA-induced PCOS rats. Peptides 2021; 145:170624. [PMID: 34375684 DOI: 10.1016/j.peptides.2021.170624] [Citation(s) in RCA: 12] [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: 06/30/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The purpose of this study was to explore the effect of dulaglutide on DHEA induced PCOS rats and its mechanism, to provide new drugs and research directions for clinical treatment of PCOS. METHODS In this study, the PCOS model was established by giving female SD rats subcutaneous injection of DHEA for 21 consecutive days. After modeling, the treatment group was injected subcutaneously with three doses of dulaglutide for 3 weeks. The model group was injected with sterile ultrapure water, and the normal group did not get any intervention. The body weight changes of rats in each group were recorded from the first day when rats received the administration of dulaglutide. Three weeks later, the rats were fasted the night after the last treatment, determined fasting insulin and fasting glucose the next day. After the rats were anesthetized by chloral hydrate, more blood was collected from the heart of the rat. The serum insulin, testosterone and sex hormone binding globulin (SHBG) levels were detected by the enzyme-linked immunoassay method. After removing the adipose tissue, the obtained rat ovary tissue was used for subsequent experimental detection, using HE staining for morphology and follicular development analysis; qRT-PCR for the detection of 3βHSD, CYP17α1, CYP19α1, and StAR gene expression in ovarian tissue; and western blotting analysis of CYP17α1, CYP19α1, StAR protein expression and insulin level to verify whether dulaglutide has a therapeutic effect on PCOS in rats. RESULTS After treated with different concentrations of dulaglutide, we found that the body weight of rats in the treatment groups were reduced. Compared with the rats in PCOS group, the serum androgen level of rats in the treatment groups was significantly decreased, and the serum sex hormone binding protein content was significantly increased, and there was statistically significant difference between these groups and PCOS group. In terms of protein expression and gene regulation, the expression of 3βHSD, CYP19α1 and StAR in the ovarian tissue of rats in treatment groups were decreased significantly after received the treatment of dulaglutide, and there was statistically significant difference between these groups and PCOS group. In addition, dulaglutide reduced the insulin content in the ovarian tissue of PCOS rats. CONCLUSION Dulaglutide may reduce the hyperandrogenemia of PCOS rats by regulating the content of serum SHBG and the expression of 3βHSD, CYP19α1, and StAR related genes and proteins, thereby inhibiting the excessive development of small follicles and the formation of cystic follicles in the ovaries of PCOS rats, thereby improving polycystic ovary in PCOS rats. In addition, dulaglutide may reduce the weight of PCOS rats, further reducing the level of high androgen in PCOS rats, and improving the morphology of their polycystic ovaries.
Collapse
Affiliation(s)
- Lu-Ming Wu
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yin-Xue Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yue Zhan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - A-Hui Liu
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Xiang Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hao-Fei Shen
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Fan Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Li-Yan Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China; Gansu Key Laboratory of Reproductive Medicine and Embryo, Gansu International Scientific and Technological Cooperation Base of Reproductive medicine transformation application, Lanzhou, China
| | - Zhong-Bin Tao
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Qing Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China; Gansu Key Laboratory of Reproductive Medicine and Embryo, Gansu International Scientific and Technological Cooperation Base of Reproductive medicine transformation application, Lanzhou, China.
| |
Collapse
|
29
|
Estradiol Regulates mRNA Levels of Estrogen Receptor Beta 4 and Beta 5 Isoforms and Modulates Human Granulosa Cell Apoptosis. Int J Mol Sci 2021; 22:ijms22095046. [PMID: 34068748 PMCID: PMC8126246 DOI: 10.3390/ijms22095046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Estrogen receptor beta (ERβ) plays a critical role in granulosa cell (GC) functions. The existence of four human ERβ splice isoforms in the ovary suggests their differential implication in 17β-estradiol (E2) actions on GC apoptosis causing follicular atresia. In this study, we investigated whether E2 can regulate ERβ isoforms expression to fine tune its apoptotic activities in human GC. For this purpose, we measured by RT-qPCR the expression of ERβ isoforms in primary culture of human granulosa cells (hGCs) collected from patients undergoing in vitro fertilization, before and after E2 exposure. Besides, we assessed the potential role of ERβ isoforms on cell growth and apoptosis after their overexpression in a human GC line (HGrC1 cells). We confirmed that ERβ1, ERβ2, ERβ4, and ERβ5 isoform mRNAs were predominant over that of ERα in hGCs, and found that E2 selectively regulates mRNA levels of ERβ4 and ERβ5 isoforms in these cells. In addition, we demonstrated that overexpression of ERβ1 and ERβ4 in HGrC1 cells increased cell apoptosis by 225% while ERβ5 or ERβ2 had no effect. Altogether, our study revealed that E2 may influence GC fate by specifically regulating the relative abundance of ERβ isoforms mRNA to modulate the balance between pro-apoptotic and non-apoptotic ERβ isoforms.
Collapse
|
30
|
Pirotta S, Joham A, Grieger JA, Tay CT, Bahri-Khomami M, Lujan M, Lim SS, Moran LJ. Obesity and the Risk of Infertility, Gestational Diabetes, and Type 2 Diabetes in Polycystic Ovary Syndrome. Semin Reprod Med 2021; 38:342-351. [PMID: 33873233 DOI: 10.1055/s-0041-1726866] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review describes the relationship between obesity and the most common reproductive (infertility) and metabolic (gestational diabetes mellitus [GDM] and type 2 diabetes mellitus [T2DM]) consequences in polycystic ovary syndrome (PCOS). It also describes the vital role of lifestyle management for PCOS. PCOS is a heterogeneous endocrine disorder common in reproductive-age women. Consensus on the exact etiological mechanisms of PCOS is unreached. Overweight or obesity is present in at least 60% of the PCOS population, but the condition occurs irrespective of BMI, with excess BMI increasing both the prevalence and severity of clinical features. Use of lifestyle therapies (nutrition, physical activity, and/or behavioral) for the prevention and management of excess weight gain, infertility, GDM, and T2DM is a vital component of best-practice PCOS care. Lifestyle management is recommended for all women with PCOS as the first-line treatment with or without medications. Due to a lack of high-quality trials demonstrating the efficacy of specific lifestyle approaches, PCOS lifestyle recommendations are as those for the general population. This review summarizes current knowledge relating to obesity and its impact on fertility, GDM, and T2DM. It also summarizes the lifestyle recommendations to best manage these conditions in women with PCOS and obesity.
Collapse
Affiliation(s)
- Stephanie Pirotta
- Health and Social Care Unit, SPHPM, Monash University, Victoria, Australia
| | - Anju Joham
- Monash Centre for Health Research and Implementation, Monash University, Victoria, Australia.,Department of Diabetes and Vascular Medicine, Monash Health, Victoria, Australia
| | - Jessica A Grieger
- Adelaide Medical School, University of Adelaide, South Australia, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Chau Tien Tay
- Monash Centre for Health Research and Implementation, Monash University, Victoria, Australia
| | - Mahnaz Bahri-Khomami
- Monash Centre for Health Research and Implementation, Monash University, Victoria, Australia
| | - Marla Lujan
- Division of Nutritional Sciences, Cornell University, New York
| | - Siew S Lim
- Monash Centre for Health Research and Implementation, Monash University, Victoria, Australia
| | - Lisa J Moran
- Monash Centre for Health Research and Implementation, Monash University, Victoria, Australia
| |
Collapse
|
31
|
Bourgneuf C, Bailbé D, Lamazière A, Dupont C, Moldes M, Farabos D, Roblot N, Gauthier C, Mathieu d'Argent E, Cohen-Tannoudji J, Monniaux D, Fève B, Movassat J, di Clemente N, Racine C. The Goto-Kakizaki rat is a spontaneous prototypical rodent model of polycystic ovary syndrome. Nat Commun 2021; 12:1064. [PMID: 33594056 PMCID: PMC7886868 DOI: 10.1038/s41467-021-21308-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is characterized by an oligo-anovulation, hyperandrogenism and polycystic ovarian morphology combined with major metabolic disturbances. However, despite the high prevalence and the human and economic consequences of this syndrome, its etiology remains unknown. In this study, we show that female Goto-Kakizaki (GK) rats, a type 2 diabetes mellitus model, encapsulate naturally all the reproductive and metabolic hallmarks of lean women with PCOS at puberty and in adulthood. The analysis of their gestation and of their fetuses demonstrates that this PCOS-like phenotype is developmentally programmed. GK rats also develop features of ovarian hyperstimulation syndrome. Lastly, a comparison between GK rats and a cohort of women with PCOS reveals a similar reproductive signature. Thus, this spontaneous rodent model of PCOS represents an original tool for the identification of the mechanisms involved in its pathogenesis and for the development of novel strategies for its treatment.
Collapse
Affiliation(s)
- Camille Bourgneuf
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Danielle Bailbé
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Antonin Lamazière
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Département PM2, Paris, France
| | - Charlotte Dupont
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | - Marthe Moldes
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Dominique Farabos
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Département PM2, Paris, France
| | - Natacha Roblot
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Camille Gauthier
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Emmanuelle Mathieu d'Argent
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | | | | | - Bruno Fève
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Service Endocrinologie, CRMR PRISIS, Paris, France
| | - Jamileh Movassat
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Nathalie di Clemente
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
- Institut Hospitalo-Universitaire ICAN, Paris, France.
- Université de Paris, Paris, France.
| |
Collapse
|
32
|
Ran Y, Yi Q, Li C. The Relationship of Anti-Mullerian Hormone in Polycystic Ovary Syndrome Patients with Different Subgroups. Diabetes Metab Syndr Obes 2021; 14:1419-1424. [PMID: 33790608 PMCID: PMC8006968 DOI: 10.2147/dmso.s299558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/05/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To explore the value of anti-Mullerian hormone (AMH) in patients with polycystic ovary syndrome (PCOS) with different phenotypes and ages, and to identify the relationship between hyperandrogenism (HA) and polycystic ovary morphology (PCOM), in a Chinese cohort. METHODS A total of 2262 women (1631 with PCOS and 631 controls) were enrolled. The serum AMH and total testosterone (TT) were analyzed, the AMH levels of each subgroup were compared, and the value of each phenotype and age group of patients with PCOS was evaluated. RESULTS The level of AMH in women with PCOS (mean±SD, 8.63±4.73 ng/mL) was higher than that in controls (5.57±3.31 ng/mL) (P<0.01). The level of AMH in the PCOM subgroup (11.19±6.4 ng/mL) was significantly higher than that in the HA subgroup (8.58±4.74 ng/mL) (P<0.01), and both were higher than that in controls (P<0.01). AMH was higher in PCOS patients than in controls, but the same values were found in subgroups of PCOS patients under 30 years old. CONCLUSION AMH changed in different subgroups of PCOS, which was the possible reason why AMH was not a diagnostic indicator. However, AMH could help to differentiate between clinical subgroups, as it was strongly related with PCOM but not with HA. AMH changed substantially with age, but was stable in PCOS patients under 30 years old.
Collapse
Affiliation(s)
- Yu Ran
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Qiang Yi
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Cong Li
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Correspondence: Cong Li Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, People’s Republic of ChinaTel +8615334506105Fax +86023 89011080 Email
| |
Collapse
|
33
|
Dilaver N, Pellatt L, Jameson E, Ogunjimi M, Bano G, Homburg R, D Mason H, Rice S. The regulation and signalling of anti-Müllerian hormone in human granulosa cells: relevance to polycystic ovary syndrome. Hum Reprod 2020; 34:2467-2479. [PMID: 31735954 DOI: 10.1093/humrep/dez214] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 09/02/2019] [Indexed: 01/14/2023] Open
Abstract
STUDY QUESTION What prevents the fall in anti-Müllerian hormone (AMH) levels in polycystic ovary syndrome (PCOS) and what are the consequences of this for follicle progression in these ovaries? SUMMARY ANSWER Exposure of granulosa cells (GCs) to high levels of androgens, equivalent to that found in PCOS, prevented the fall in AMH and was associated with dysregulated AMH-SMAD signalling leading to stalled follicle progression in PCOS. WHAT IS KNOWN ALREADY In normal ovaries, AMH exerts an inhibitory role on antral follicle development and a fall in AMH levels is a prerequisite for ovulation. Levels of AMH are high in PCOS, contributing to the dysregulated follicle growth that is a common cause of anovulatory infertility in these women. STUDY DESIGN, SIZE, DURATION Human KGN-GC (the cell line that corresponds to immature GC from smaller antral follicles (AF)) were cultured with a range of doses of various androgens to determine the effects on AMH production. KGN-GC were also treated with PHTPP (an oestrogen receptor β (ERβ) antagonist) to examine the relationship between AMH expression and the ratio of ERα:ERβ. The differential dose-related effect of AMH on gene expression and SMAD signalling was investigated in human granulosa-luteal cells (hGLC) from women with normal ovaries, with polycystic ovarian morphology (PCOM) and with PCOS. KGN-GC were also cultured for a prolonged period with AMH at different doses to assess the effect on cell proliferation and viability. PARTICIPANTS/MATERIALS, SETTING, METHODS AMH protein production by cells exposed to androgens was measured by ELISA. The effect of PHTPP on the mRNA expression levels of AMH, ERα and ERβ was assessed by real-time quantitative PCR (qPCR). The influence of AMH on the relative mRNA expression levels of aromatase, AMH and its receptor AMHRII, and the FSH and LH receptor (FSHR and LHR) in control, PCOM and PCOS hGLCs was quantified by qPCR. Western blotting was used to assess changes in levels of SMAD proteins (pSMAD-1/5/8; SMAD-4; SMAD-6 and SMAD-7) after exposure of hGLCs from healthy women and women with PCOS to AMH. The ApoTox-Glo Triplex assay was used to evaluate the effect of AMH on cell viability, cytotoxicity and apoptosis. MAIN RESULTS AND THE ROLE OF CHANCE Testosterone reduced AMH protein secreted from KGN-GC at 10-9-10-7 M (P < 0.05; P < 0.005, multiple uncorrected comparisons Fishers least squares difference), but at equivalent hyperandrogenemic levels no change was seen in AMH levels. 5α-DHT produced a significant dose-related increase in AMH protein secreted into the media (P = 0.022, ANOVA). Increasing the mRNA ratio of ERα:ERβ produced a corresponding increase in AMH mRNA expression (P = 0.015, two-way ANOVA). AMH increased mRNA levels of aromatase (P < 0.05, one-way ANOVA) and FSHR (P < 0.0001, one-way ANOVA) in hGLCs from women with PCOM, but not from normal cells or PCOS (normal n = 7, PCOM n = 5, PCOS n = 4). In contrast to hGLCs from ovulatory ovaries, in PCOS AMH reduced protein levels (cell content) of stimulatory pSMAD-1/5/8 and SMAD-4 but increased inhibitory SMAD-6 and -7 (P < 0.05, normal n = 6, PCOS n = 3). AMH at 20 and 50 ng/ml decreased KGN-GC cell proliferation but not viability after 8 days of treatment (P < 0.005, two-way ANOVA). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Luteinised GC from women undergoing IVF have a relatively low expression of AMH/AMHRII but advantageously continue to display responses inherent to the ovarian morphology from which they are collected. To compensate, we also utilised the KGN cell line which has been characterised to be at a developmental stage close to that of immature GC. The lack of flutamide influence on testosterone effects is not in itself sufficient evidence to conclude that the effect on AMH is mediated via conversion to oestrogen, and the effect of aromatase inhibitors or oestrogen-specific inhibitors should be tested. The effect of flutamide was tested on testosterone but not DHT. WIDER IMPLICATIONS OF THE FINDINGS Normal folliculogenesis and ovulation are dependent on the timely reduction in AMH production from GC at the time of follicle selection. Our findings reveal for the first time that theca-derived androgens may play a role in this model but that this inhibitory action is lost at levels of androgens equivalent to those seen in PCOS. The AMH decline may either be a direct effect of androgens or an indirect one via conversion to oestradiol and acting through the upregulation of ERα, which is known to stimulate the AMH promoter. Interestingly, the ability of GCs to respond to this continually elevated AMH level appears to be reduced in cells from women with PCOS due to an adaptive alteration in the SMAD signalling pathway and lower expression of AMHRII, indicating a form of 'AMH resistance'. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the Thomas Addison Scholarship, St Georges Hospital Trust. The authors report no conflict of interest in this work and have nothing to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Nafi Dilaver
- Cell Biology and Genetics Research Centre, St George's University of London, London SW17 0RE, UK.,Academic Foundation Programme, Imperial College London, Charing Cross Hospital, London W6 8RF, UK
| | - Laura Pellatt
- Cell Biology and Genetics Research Centre, St George's University of London, London SW17 0RE, UK.,Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent, ME4 4TB, UK
| | - Ella Jameson
- Biomedical Science Undergraduate Programme, St George's University of London, London SW17 0RE, UK
| | - Michael Ogunjimi
- Biomedical Science Undergraduate Programme, St George's University of London, London SW17 0RE, UK
| | - Gul Bano
- Thomas Addison Endocrine Unit, St George's Hospital, Cranmer Terrace, London SW17 0RE, UK
| | - Roy Homburg
- Homerton Fertility Unit, Homerton University Hospital, Homerton Row, London, UK
| | - Helen D Mason
- Cell Biology and Genetics Research Centre, St George's University of London, London SW17 0RE, UK
| | - Suman Rice
- Cell Biology and Genetics Research Centre, St George's University of London, London SW17 0RE, UK
| |
Collapse
|
34
|
Estienne A, Jarrier P, Staub C, Venturi E, Le Vern Y, Clemente N, Monniaux D, Monget P. Anti-Müllerian hormone production in the ovary: a comparative study in bovine and porcine granulosa cells†. Biol Reprod 2020; 103:572-582. [PMID: 32432313 DOI: 10.1093/biolre/ioaa077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/04/2020] [Indexed: 12/23/2022] Open
Abstract
In this study, we aimed to determine the origin of the difference, in terms of anti-Müllerian hormone production, existing between the bovine and porcine ovaries. We first confirmed by quantitative real-time-Polymerase-Chain Reaction, ELISA assay and immunohistochemistry that anti-Müllerian hormone mRNA and protein production are very low in porcine ovarian growing follicles compared to bovine ones. We then have transfected porcine and bovine granulosa cells with vectors containing the luciferase gene driven by the porcine or the bovine anti-Müllerian hormone promoter. These transfection experiments showed that the porcine anti-Müllerian hormone promoter is less active and less responsive to bone morphogenetic protein stimulations than the bovine promoter in both porcine and bovine cells. Moreover, bovine but not porcine granulosa cells were responsive to bone morphogenetic protein stimulation after transfection of a plasmidic construction including a strong response element to the bone morphogenetic proteins (12 repetitions of the GCCG sequence) upstream of the luciferase reporter gene. We also showed that SMAD6, an inhibitor of the SMAD1-5-8 pathway, is strongly expressed in porcine compared to the bovine granulosa cells. Overall, these results suggest that the low expression of anti-Müllerian hormone in porcine growing follicles is due to both a lack of activity/sensitivity of the porcine anti-Müllerian hormone promoter, and to the lack of responsiveness of porcine granulosa cells to bone morphogenetic protein signaling, potentially due to an overexpression of SMAD6 compared to bovine granulosa cells. We propose that the low levels of anti-Müllerian hormone in the pig would explain the poly-ovulatory phenotype in this species.
Collapse
Affiliation(s)
- Anthony Estienne
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Françis du Cheval et de l'Equitation (IFCE), Université de Tours, Tours, France
| | - Peggy Jarrier
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Françis du Cheval et de l'Equitation (IFCE), Université de Tours, Tours, France
| | - Christophe Staub
- Physiologie Animale et Systèmes d'Elevage, Unité Expérimentale de Physiologie Animale de l'Orfrasière (UEPAO), Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Unité Expérimentale (UE) 1297, Nouzilly, France
| | - Eric Venturi
- Physiologie Animale et Systèmes d'Elevage, Unité Expérimentale de Physiologie Animale de l'Orfrasière (UEPAO), Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Unité Expérimentale (UE) 1297, Nouzilly, France
| | - Yves Le Vern
- Infectiologie, Santé Publique (ISP), Unité Mixte de Recherche (UMR) 1297, Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Université de Tours, Tours, France
| | - Nathalie Clemente
- Sorbonne Université, Insitut National de la Santé et de la Recherche Médicale (INSERM), Centre de Recherche Saint-Antoine, Paris, France
| | - Danielle Monniaux
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Françis du Cheval et de l'Equitation (IFCE), Université de Tours, Tours, France
| | - Philippe Monget
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique et de l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Françis du Cheval et de l'Equitation (IFCE), Université de Tours, Tours, France
| |
Collapse
|
35
|
Trent M, Gordon CM. Diagnosis and Management of Polycystic Ovary Syndrome in Adolescents. Pediatrics 2020; 145:S210-S218. [PMID: 32358213 DOI: 10.1542/peds.2019-2056j] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2020] [Indexed: 11/24/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common female reproductive disorder that often manifests during adolescence and is associated with disruptions in health-related quality of life. Prompt evaluation and clinical support after diagnosis may prevent associated complications and optimize overall health management. This article incorporates the most recent evidence and consensus guidelines to provide an updated review of the pathogenesis, clinical presentation, diagnostic evaluation, and management strategies for adolescents with this complex condition. We will review the recent international guidelines on PCOS; because the diagnosis of PCOS remains controversial, management of this condition is inconsistent. In 2019, PCOS remains a common, yet neglected, condition, in part, because of the lack of agreement around both diagnosis and management.
Collapse
Affiliation(s)
- Maria Trent
- Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland; and
| | - Catherine M Gordon
- Division of Adolescent/Young Adult Medicine, Harvard Medical School, Harvard University and Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
36
|
Dewailly D, Barbotin AL, Dumont A, Catteau-Jonard S, Robin G. Role of Anti-Müllerian Hormone in the Pathogenesis of Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2020; 11:641. [PMID: 33013710 PMCID: PMC7509053 DOI: 10.3389/fendo.2020.00641] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
Besides its interest for diagnosis, the finding of an elevated serum AMH level in PCOS has open major pathophysiological issues. This review addresses the three most important issues: 1- the role of AMH in the disturbed folliculogenesis of PCOS; 2- the role of AMH in the gonadotropin dysregulation of PCOS and 3- the role of AMH in the trans-generational transmission of PCOS. For each of those issues, the clinical and experimental evidences currently available are discussed and pathophysiological hypothesis are proposed.
Collapse
Affiliation(s)
- Didier Dewailly
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Lille, France
- *Correspondence: Didier Dewailly
| | - Anne-Laure Barbotin
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Lille, France
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Hôpital Jeanne de Flandre, Lille, France
| | - Agathe Dumont
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France
- CHU Lille, Service d'Assistance Médicale à la Procréation et Préservation de la Fertilité, Hôpital Jeanne de Flandre, Lille, France
| | - Sophie Catteau-Jonard
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Lille, France
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France
| | - Geoffroy Robin
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France
- CHU Lille, Service d'Assistance Médicale à la Procréation et Préservation de la Fertilité, Hôpital Jeanne de Flandre, Lille, France
- Lille University, EA 4308 “Gametogenesis and Gamete Quality”, Lille, France
| |
Collapse
|
37
|
Olivier LS, Evliyaoglu O, Weiskirchen R, van Helden J. Investigation of soluble anti-Müllerian hormone receptor type 2 as a biomarker for diagnosis of female fertility disorders. Reprod Biomed Online 2019; 39:1017-1025. [PMID: 31727499 DOI: 10.1016/j.rbmo.2019.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 10/26/2022]
Abstract
RESEARCH QUESTION The ectodomain of the anti-Müllerian hormone (AMH) type 2 receptor is shed by proteases under certain conditions, which makes it measurable in the blood. The aim of this study was to identify correlations of soluble anti-Müllerian hormone receptor type 2 (sAMHR2) with other sex hormone concentrations and to assess whether sAMHR2 may serve as a new biomarker in fertility disorders. DESIGN In a retrospective cross-sectional study of women (n = 186) with different gynaecological-endocrinological disorders, mixed-effect models were used to analyse the correlation with established diagnostic hormone tests. Receiver operating characteristic curve analysis was performed to assess the diagnostic performance. RESULTS There was a strong correlation of sAMHR2 with LH (r = 0.898) and FSH (r = 0.846) and a moderate correlation of AMH with testosterone (r = 0.666) and androstenedione (r = 0.696) (all P < 0.001). In diagnoses of polycystic ovary syndrome (PCOS), AMH showed the best performance (area under the curve [AUC] 0.981, cut-off 4 ng/ml) with 96% sensitivity and 94% specificity. sAMHR2 concentrations and sAMHR2/AMH ratios were elevated in women with ovarian insufficiency, compared with all other study groups, including post-menopausal women on hormone replacement therapy. Highest sensitivity and specificity (100% and 98.2%, respectively) were achieved with sAMHR2/AMH ratio for the diagnosis of post-menopausal status (cut-off 68.85). The sAMHR2/AMH ratio (AUC 0.997) had a better performance than sAMHR2 (AUC 0.947), FSH (AUC 0.989) and LH (AUC 0.967). CONCLUSIONS The sAMHR2/AMH ratio may serve as a useful biomarker for infertility diagnostics to identify post-menopausal women.
Collapse
Affiliation(s)
- Lena Sophie Olivier
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, Germany
| | - Osman Evliyaoglu
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, Germany.
| | - Josef van Helden
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Germany.
| |
Collapse
|
38
|
Polycystic ovarian syndrome: Correlation between hyperandrogenism, insulin resistance and obesity. Clin Chim Acta 2019; 502:214-221. [PMID: 31733195 DOI: 10.1016/j.cca.2019.11.003] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex and heterogeneous endocrine disease characterized by clinical or laboratorial hyperandrogenism, oligo-anovulation and metabolic abnormalities, including insulin resistance, excessive weight or obesity, type II diabetes, dyslipidemia and an increased risk of cardiovascular disease. The most significant clinical manifestation of PCOS is hyperandrogenism. Excess androgen profoundly affects granulosa cell function and follicular development via complex mechanisms that lead to obesity and insulin resistance. Most PCOS patients with hyperandrogenism have steroid secretion defects that result in abnormal folliculogenesis and failed dominant follicle selection. Hyperandrogenism induces obesity, hairy, acne, and androgenetic alopecia. These symptoms can bring great psychological stress to women. Drugs such as combined oral contraceptive pills, metformin, pioglitazone and low-dose spironolactone help improve pregnancy rates by decreasing androgen levels in vivo. Notably, PCOS is heterogeneous, and hyperandrogenism is not the only pathogenic factor. Obesity and insulin resistance aggravate the symptoms of hyperandrogenism, forming a vicious cycle that promotes PCOS development. Although numerous studies have been conducted, the definitive pathogenic mechanisms of PCOS remain uncertain. This review summarizes and discusses previous and recent findings regarding the relationship between hyperandrogenism, insulin resistance, obesity and PCOS.
Collapse
|
39
|
Li Y, Gao D, Xu T, Adur MK, Zhang L, Luo L, Zhu T, Tong X, Zhang D, Wang Y, Ning W, Qi X, Cao Z, Zhang Y. Anti-Müllerian hormone inhibits luteinizing hormone-induced androstenedione synthesis in porcine theca cells. Theriogenology 2019; 142:421-432. [PMID: 31711705 DOI: 10.1016/j.theriogenology.2019.10.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 11/28/2022]
Abstract
AMH (Anti-Müllerian Hormone) is involved in the regulation of follicle growth initiation and inhibits FSH-induced aromatase expression and estrogen production in granulosa cells. However, the function of AMH in steroidogenesis by theca cells remains unclear. The aim of this study is to investigate the role of AMH as a regulator of the basal and stimulated steroid production by pig granulosa cells (pGCs) and theca cells (pTCs). PGCs and pTCs were incubated with hormones AMH, LH (luteinizing hormone), FSH (follicle stimulating hormone), individually or in combination. The expression of CYP19A1, HSD3B1, CYP11A1, LHCGR, and CYP17A1 mRNA were evaluated by quantitative reverse transcriptase PCR. In pGCs, 10 ng/mL AMH significantly decreased the FSH-stimulated effect on FSHR and CYP19A1 expression and estradiol production. In pTCs, LH treatment significantly increased the expression of HSD3B1, CYP11A1, LHCGR, and androstenedione or progesterone production (P < 0.05). Additionally, 10 ng/mL AMH also significantly decreased the LH-stimulated effects on the expression of HSD3B1, CYP11A1, CYP17A1, LHCGR and androstenedione production. Transfection with siAMHR2-I abolished the suppressive effects of AMH on LH-induced HSD3B1 expression and androstenedione production. Taken together, these results demonstrate that AMH is involved in FSH induced estradiol production in pGCs and LH induced androstenedione production in pTCs by regulating the steroidogenesis pathway.
Collapse
Affiliation(s)
- Yunsheng Li
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Di Gao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Tengteng Xu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Malavika K Adur
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Ling Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Lei Luo
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Ting Zhu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xu Tong
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Dandan Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Yiqing Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Wei Ning
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xin Qi
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Zubing Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
40
|
Huang CC, Chen MJ, Lan CW, Wu CE, Huang MC, Kuo HC, Ho HN. Hyperactive CREB signaling pathway involved in the pathogenesis of polycystic ovarian syndrome revealed by patient-specific induced pluripotent stem cell modeling. Fertil Steril 2019; 112:594-607.e12. [PMID: 31277818 DOI: 10.1016/j.fertnstert.2019.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To study whether and how the pathogenesis of polycystic ovarian syndrome (PCOS) is related to epigenetic aberrations. DESIGN A case-control experimental study. SETTING Tertiary university hospital. PATIENT(S) Eighteen patients with PCOS and ten non-PCOS control subjects. INTERVENTIONS(S) Patient-specific induced pluripotent stem cells (iPSCs) were obtained from skin fibroblasts through the application of nonviral episomal reprogramming and were differentiated into ovarian granulosa cells (GCs) with the use of a cocktail of growth factors. Primary ovarian GCs were collected during transvaginal oocyte retrieval surgery. MAIN OUTCOME MEASURE(S) Characterization and functional validation of iPSC-derived GCs were conducted. Whole-genomic DNA methylation profiles in women with and without PCOS in both iPSC-derived GCs and primary adult GCs were analyzed with the use of the Illumina 850K MethylationEPIC Beadchip. RESULT(S) The iPSC-derived GCs successfully expressed GC-associated genes and aromatase activity after differentiation. Whole-genomic DNA methylation analysis of the iPSC-derived GCs and adult GCs both revealed a hyperactive CREB signaling pathway in the PCOS group compared with the control group. The expression of CREB-binding protein (CBP) mRNA was significantly higher in the iPSC-derived GCs in the PCOS group, and the expression of CBP protein was also significantly higher in the primary GCs from women with PCOS. CONCLUSION(S) The combination of DNA methylomic analysis in primary adult GCs and iPSC-derived GCs showed that a preserved persistent hyperactivation of the CREB signaling pathway might be involved in the pathogenesis of PCOS. These results could have implications on the early developmental origin, inheritance nature, and environmental interaction effects of this disease.
Collapse
Affiliation(s)
- Chu-Chun Huang
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan.
| | - Chen-Wei Lan
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan
| | - Chia-Eng Wu
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan
| | - Mei-Chi Huang
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan
| | - Hung-Chih Kuo
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hong-Nerng Ho
- Department of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
41
|
Li Y, Chen C, Ma Y, Xiao J, Luo G, Li Y, Wu D. Multi-system reproductive metabolic disorder: significance for the pathogenesis and therapy of polycystic ovary syndrome (PCOS). Life Sci 2019; 228:167-175. [PMID: 31029778 DOI: 10.1016/j.lfs.2019.04.046] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/02/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022]
Abstract
Polycystic ovary syndrome (PCOS), a multisystem disease, is a major reason for female infertility around the world. It is no longer considered simply as a disease of ovary. Now researchers growing awareness of the multisystem features of this disease. PCOS has a higher relationship with metabolic disturbance and hypothalamic-pituitary-ovarian axis (HPOA) function disorders. This syndrome results in hyperandrogenemia (HA), hyperinsulinemia/insulin resistance (IR), increased estrone, luteinizing hormone (LH) and follicle-stimulating hormone (FSH) ratio imbalance, infertility, cardiovascular diseases, endometrial dysfunction, obesity, and including a litany of other health issues. Furthermore, PCOS has been garnered in recent times. Interventions like metformin, orlistat, hormonal contraceptives, GLP1 agonists, and VitD have been applied to ameliorate or reverse the pathological characterization of PCOS. Moreover, drug-combined therapy of PCOS is superior to single drug administration. This review will focus on the recent progress in pathogenesis and therapy of PCOS.
Collapse
Affiliation(s)
- Yan Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, PR China; Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan 410078, PR China
| | - Changye Chen
- Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang 421001, PR China
| | - Yan Ma
- Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang 421001, PR China
| | - Jiao Xiao
- Department of Endocrinology, The Affiliated Nanhua Hospital, University of South China, Hengyang 421002, PR China
| | - Guifang Luo
- Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang 421001, PR China
| | - Yukun Li
- Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, PR China.
| | - Daichao Wu
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, PR China; University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA.
| |
Collapse
|
42
|
Efthymiadou A, Bogiatzidou M, Kritikou D, Chrysis D. Anti-Müllerian Hormone in Girls with Premature Adrenarche: The Impact of Polycystic Ovary Syndrome History in their Mothers. J Pediatr 2019; 205:190-194. [PMID: 30529136 DOI: 10.1016/j.jpeds.2018.09.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/14/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVES To assess whether the serum levels of anti-Müllerian hormone (AMH) are increased in girls with premature adrenarche because they are at a higher risk of developing polycystic ovary syndrome (PCOS) later in life. STUDY DESIGN We measured serum levels of AMH, dehydroepiandrosterone sulfate (DHEAS), testosterone, sex hormone binding globulin, androstenedione, and 17-hyroxyprogesterone in 89 girls with premature adrenarche aged 6.98 ± 1.60 years, and in 55 prepubertal normal girls aged 6.78 ± 1.60 years. RESULTS AMH was significantly higher in girls with premature adrenarche (2.95 ± 1.20 ng/mL) compared with normal prepubertal girls (2.00 ± 0.95 ng/mL; P < .001), whereas their body mass index SD score was similar (P > .05). DHEAS, testosterone, and androstenedione were increased in premature adrenarche, whereas sex hormone binding globulin was decreased in girls with premature adrenarche. Among the 89 girls with premature adrenarche, 33 were daughters of mothers with a positive history of PCOS, whereas the mothers of the remaining 56 girls with premature adrenarche had a negative history of PCOS. The girls with a mother with a positive history of PCOS had significantly higher AMH serum levels compared with girls with a mother with a negative history of PCOS (3.37 ± 1.72 ng/mL vs 2.70 ± 1.25 ng/mL; P < .05) with no differences in testosterone, DHEAS, androstenedione, and sex hormone binding globulin. The serum concentration of AMH was only positively related to androstenedione (r = 0.538; P < .0001). CONCLUSIONS Girls with premature adrenarche, especially those from mothers with a history of PCOS, could have a higher risk of developing PCOS later in life because they have increased serum AMH.
Collapse
Affiliation(s)
- Alexandra Efthymiadou
- Division of Endocrinology Department of Pediatrics, Medical School, University of Patras, Patra, Greece
| | - Maria Bogiatzidou
- Division of Endocrinology Department of Pediatrics, Medical School, University of Patras, Patra, Greece
| | - Dimitra Kritikou
- Division of Endocrinology Department of Pediatrics, Medical School, University of Patras, Patra, Greece
| | - Dionisios Chrysis
- Division of Endocrinology Department of Pediatrics, Medical School, University of Patras, Patra, Greece.
| |
Collapse
|
43
|
Piltonen TT, Giacobini P, Edvinsson Å, Hustad S, Lager S, Morin-Papunen L, Tapanainen JS, Sundström-Poromaa I, Arffman RK. Circulating antimüllerian hormone and steroid hormone levels remain high in pregnant women with polycystic ovary syndrome at term. Fertil Steril 2019; 111:588-596.e1. [PMID: 30630591 DOI: 10.1016/j.fertnstert.2018.11.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/05/2018] [Accepted: 11/19/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate plasma antimüllerian hormone (AMH) concentration and its relation to steroid hormone levels in pregnant women with polycystic ovary syndrome (PCOS) and controls at term. DESIGN Case-control study. SETTING University-affiliated hospital. PATIENT(S) A total of 74 pregnant women at term: 25 women with PCOS (aged 31.6 ± 3.9 years [mean ± standard deviation], body mass index 24.0 ± 3.9 kg/m2, mean gestational length 279 ± 9 days) and 49 controls (aged 31.7 ± 3.3 years, body mass index 24.0 ± 3.3 kg/m2, mean gestational length 281 ± 9 days). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Plasma AMH and steroid hormone levels. RESULT(S) Antimüllerian hormone, T, and androstenedione levels were higher in women with PCOS at term compared with controls, whereas estrogen and P levels were similar. The differences were pronounced in women carrying a female fetus. Testosterone and AMH levels correlated positively in both groups, but E2 levels only in women with PCOS. CONCLUSION(S) Pregnant women with PCOS present with elevated AMH and androgen levels even at term, suggesting a hormonal imbalance during PCOS pregnancy. Differences were detected especially in pregnancies with a female fetus, raising the question of whether female pregnancies are more susceptible to AMH and steroid hormone actions.
Collapse
Affiliation(s)
- Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.
| | - Paolo Giacobini
- Jean-Pierre Aubert Research Center, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Institut National de la Santé et de la Recherche Médicale, Lille, France; Fédération Hospitalière Universitaire, School of Medicine, University of Lille, Lille, France
| | - Åsa Edvinsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Steinar Hustad
- Department of Clinical Science, University of Bergen, Bergen, Norway; Core Facility for Metabolomics, University of Bergen, Bergen, Norway
| | - Susanne Lager
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland; Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
44
|
Dumont A, Robin G, Dewailly D. Anti-müllerian hormone in the pathophysiology and diagnosis of polycystic ovarian syndrome. Curr Opin Endocrinol Diabetes Obes 2018; 25:377-384. [PMID: 30299432 DOI: 10.1097/med.0000000000000445] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Polycystic ovarian syndrome (PCOS) is the most common cause of chronic anovulation and hyperandrogenism in young women and represents a true public health concern and an economic burden. RECENT FINDINGS The pathophysiology of PCOS is still not fully understood, but progresses have been made and the relationships between anti mullerian hormone (AMH), follicle stimulating hormone, luteinizing hormone, E2 and androgens have been explored. The follicle excess plays a central role in the syndrome and AMH is definitively a major component of this phenomena. SUMMARY The aim of this chapter is to present the recent work studying the role of AMH in the pathophysiology of PCOS and to discuss the improvement that serum AMH assay brings in the diagnosis of PCOS.
Collapse
Affiliation(s)
- Agathe Dumont
- Service de Gynécologie Endocrinienne et de Médecine de la Reproduction, Hôpital Jeanne de Flandre, CHRU, Lille, France
| | | | | |
Collapse
|
45
|
Garrel G, Denoyelle C, L'Hôte D, Picard JY, Teixeira J, Kaiser UB, Laverrière JN, Cohen-Tannoudji J. GnRH Transactivates Human AMH Receptor Gene via Egr1 and FOXO1 in Gonadotrope Cells. Neuroendocrinology 2018; 108:65-83. [PMID: 30368511 DOI: 10.1159/000494890] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/26/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND/OBJECTIVES Anti-Müllerian hormone (AMH) signaling is critical for sexual differentiation and gonadal function. AMH receptor type 2 (AMHR2) is expressed in extragonadal sites such as brain, and pituitary and emerging evidence indicates that AMH biological action is much broader than initially thought. We recently reported that AMH signaling enhances follicle-stimulating hormone synthesis in pituitary gonadotrope cells. However, mechanisms regulating AMHR2 expression in these extragonadal sites remain to be explored. METHOD/RESULTS Here, we demonstrated in perifused murine LβT2 gonadotrope cells that Amhr2 expression is differentially regulated by GnRH pulse frequency with an induction under high GnRH pulsatility. Furthermore, we showed that GnRH transactivates the human AMHR2 promoter in LβT2 cells. Successive deletions of the promoter revealed the importance of a short proximal region (-53/-37 bp) containing an Egr1 binding site. Using site-directed mutagenesis of Egr1 motif and siRNA mediated-knockdown of Egr1, we demonstrated that Egr1 mediates basal and GnRH-dependent activity of the promoter, identifying Egr1 as a new transcription factor controlling hAMHR2 expression. We also showed that SF1 and β-catenin are required for basal promoter activity and demonstrated that both factors contribute to the GnRH stimulatory effect, independently of their respective binding sites. Furthermore, using a constitutively active mutant of FOXO1, we identified FOXO1 as a negative regulator of basal and GnRH-dependent AMHR2 expression in gonadotrope cells. CONCLUSIONS This study identifies GnRH as a regulator of human AMHR2 expression, further highlighting the importance of AMH signaling in the regulation of gonadotrope function.
Collapse
Affiliation(s)
- Ghislaine Garrel
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris, France
| | - Chantal Denoyelle
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris, France
| | - David L'Hôte
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris, France
| | - Jean-Yves Picard
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris, France
| | - Jose Teixeira
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jean-Noël Laverrière
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris, France
| | - Joëlle Cohen-Tannoudji
- Physiologie de l'axe gonadotrope U1133, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Biologie Fonctionnelle et Adaptative UMR 8251, Sorbonne Paris Cité, Université Paris-Diderot, Paris,
| |
Collapse
|
46
|
Xue P, Wang Z, Fu X, Wang J, Punchhi G, Wolfe A, Wu S. A Hyperandrogenic Mouse Model to Study Polycystic Ovary Syndrome. J Vis Exp 2018. [PMID: 30346398 DOI: 10.3791/58379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Hyperandrogenemia plays a critical role in reproductive and metabolic function in females and is the hallmark of polycystic ovary syndrome. Developing a lean PCOS-like mouse model that mimics women with PCOS is clinically meaningful. In this protocol, we describe such a model. By inserting a 4 mm length of DHT (dihydrotestosterone) crystal powder pellet (total length of pellet is 8 mm), and replacing it monthly, we are able to produce a PCOS-like mouse model with serum DHT levels 2 fold higher than mice not implanted with DHT (no-DHT). We observed reproductive and metabolic dysfunction without changing body weight and body composition. While exhibiting a high degree of infertility, a small subset of these PCOS-like female mice can get pregnant and their offspring show delayed puberty and increased testosterone as adults. This PCOS-like lean mouse model is a useful tool to study the pathophysiology of PCOS and the offspring from these PCOS-like dams.
Collapse
Affiliation(s)
- Ping Xue
- Department of Pediatrics, Johns Hopkins University School of Medicine
| | - Zhiqiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine
| | - Xiaomin Fu
- Department of Pediatrics, Johns Hopkins University School of Medicine; Department of Health, Beijing Military General Hospital
| | - Junjiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine; Southern Medical University
| | - Gopika Punchhi
- Department of Pediatrics, Johns Hopkins University School of Medicine
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine; Department of Molecular and Cellular Physiology, Johns Hopkins University School of Medicine
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine; Department of Molecular and Cellular Physiology, Johns Hopkins University School of Medicine; Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine;
| |
Collapse
|