1
|
Naqvi I, Bandyopadhyay A, Panda A, Hareramadas B. Polycystic Ovarian Syndrome: A Review of Multi-omics Analyses. Reprod Sci 2025; 32:618-646. [PMID: 39875694 DOI: 10.1007/s43032-025-01789-8] [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: 03/27/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Polycystic Ovary Syndrome (PCOS) is among the most prevalent endocrinological abnormalities of young females, posing a grave public health challenge to the society. The objective of the present literature review is to analyze the enormous amount of information available by way of numerous multi-omic studies, and to explore a meaningful relationship between various factors such as genetic, proteomic, environmental etc. to understand the multifactorial metabolic disorder in a proper manner. Detailed literature search was done in various science article repositories and biomedical databases such as PubMed, Google Scholar, BioMed Central, Embase etc. by using several keywords in whole gamut of combinations. PCOS is a heritable disease. It manifests as a result of a combination of several intricately inter-linked symptoms such as anovulation, obesity, type II diabetes, hyperandrogenism, polycystic ovaries etc., the last one being the main manifestation of the disease, thus leading to infertility among several other complications. Such a multifactorial metabolic disorder with extreme symptomatic heterogeneity cannot be fully explained solely based on symptoms or genetic variations; thus, giving some space of thought to other factors such as epigenetic, microbiomic factors etc. playing a role in the causation of the disease. The present scientific survey of literature extensively reviews various aspects of PCOS by critically looking into the vast multi-omic data, and concluded with suggesting treatment options as well as lifestyle changes required to deal with the psychological/ emotional impacts of the condition on affected women.
Collapse
Affiliation(s)
- Ilmas Naqvi
- Department of Zoology, Zakir Husain Delhi College (University of Delhi), J.L.N. Marg, New Delhi, 110002, India
| | | | - Amisha Panda
- Lab. No. 115, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - B Hareramadas
- Department of Zoology, Zakir Husain Delhi College (University of Delhi), J.L.N. Marg, New Delhi, 110002, India.
| |
Collapse
|
2
|
Włodarczyk M, Ciebiera M, Nowicka G, Łoziński T, Ali M, Al-Hendy A. Epigallocatechin Gallate for the Treatment of Benign and Malignant Gynecological Diseases-Focus on Epigenetic Mechanisms. Nutrients 2024; 16:559. [PMID: 38398883 PMCID: PMC10893337 DOI: 10.3390/nu16040559] [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: 01/09/2024] [Revised: 02/10/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
The most common malignant gynecologic diseases are cervical, uterine, ovarian, vaginal, and vulvar cancer. Among them, ovarian cancer causes more deaths than any other cancer of the female reproductive system. A great number of women suffer from endometriosis, uterine fibroids (UFs), adenomyosis, dysmenorrhea, and polycystic ovary syndrome (PCOS), which are widespread benign health problems causing troublesome and painful symptoms and significantly impairing the quality of life of affected women, and they are some of the main causes of infertility. In addition to the available surgical and pharmacological options, the effects of supporting standard treatment with naturally occurring compounds, mainly polyphenols, are being studied. Catechins are responsible for the majority of potential health benefits attributed to green tea consumption. Epigallocatechin gallate (EGCG) is considered a non-toxic, natural compound with potential anticancer properties. Antioxidant action is its most common function, but attention is also drawn to its participation in cell division inhibition, apoptosis stimulation and epigenetic regulation. In this narrative review, we describe the role of EGCG consumption in preventing the development of benign reproductive disorders such as UF, endometriosis, and PCOS, as well as malignant gynecologic conditions. We discuss possible epigenetic mechanisms that may be related to the action of EGCG.
Collapse
Affiliation(s)
- Marta Włodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland;
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
- Development and Research Center of Non-Invasive Therapies, Pro-Familia Hospital, 35-302 Rzeszów, Poland
| | - Grażyna Nowicka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Tomasz Łoziński
- Department of Obstetrics and Gynecology, Pro-Familia Hospital, 35-302 Rzeszow, Poland;
- Department of Gynecology and Obstetrics, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (M.A.); (A.A.-H.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (M.A.); (A.A.-H.)
| |
Collapse
|
3
|
Miranda AG, Seneda MM, Faustino LR. DNA methylation associated with polycystic ovary syndrome: a systematic review. Arch Gynecol Obstet 2024; 309:373-383. [PMID: 37119419 DOI: 10.1007/s00404-023-07025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 03/08/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is an endocrine metabolic disease that affects women of reproductive age and is one of the main causes of anovulatory infertility. However, the cause of PCOS is yet fully understood, and genetic factors play an important role in its etiology. In this study, we reviewed the main genes involved in the etiology of PCOS and the influence of DNA methylation, aiming to answer the study´s guiding question: 'What is the influence of DNA methylation on the main genes involved in PCOS?'. METHODS We used the MEDLINE database, and inclusion criteria (primary and original articles, written in English, found through our entry terms) and exclusion criteria (literature reviews and articles that used animals to perform the experiments and that focused in other epigenetics mechanism without being DNA methylation) were applied. RESULTS Twenty-three scientific articles, from a total of 43 articles read in full, were chosen for this study. Eighteen studies confirmed DNA methylation associated with PCOS. CONCLUSION The most relevant genes related to PCOS were INSR, LHCGR, and RAB5B, which may be epigenetically altered in DNA, with the first two genes hypomethylated and the last hypermethylated. The epigenetic changes presented in the genes related to PCOS or their promoters were only at the CpG sites.
Collapse
Affiliation(s)
- Arícia Gomes Miranda
- Curso de Medicina, Campus Ministro Reis Velloso, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, Piauí, Brazil
| | - Marcelo Marcondes Seneda
- Laboratório de Reprodução Animal, Centro de Ciências Agrárias, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Luciana Rocha Faustino
- Curso de Medicina, Campus Ministro Reis Velloso, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, Piauí, Brazil.
| |
Collapse
|
4
|
Zhang F, Ding Y, Zhang B, He M, Wang Z, Lu C, Kang Y. Analysis of Methylome, Transcriptome, and Lipid Metabolites to Understand the Molecular Abnormalities in Polycystic Ovary Syndrome. Diabetes Metab Syndr Obes 2023; 16:2745-2763. [PMID: 37720421 PMCID: PMC10503565 DOI: 10.2147/dmso.s421947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/30/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose This study aimed to identify differentially methylated genes (DMGs) and differentially expressed genes (DEGs) to investigate new biomarkers for the diagnosis and treatment of polycystic ovary syndrome (PCOS). Methods To explore the potential biomarkers of PCOS diagnosis and treatment, we performed methyl-binding domain sequencing (MBD-seq) and RNA sequencing (RNA-seq) on ovarian granulosa cells (GCs) from PCOS patients and healthy controls. MBD-seq was also performed on the ovarian tissue of constructed prenatally androgenized (PNA) mice. Differential methylation and expression analysis were implemented to identify DMGs and DEGs, respectively. The identified gene was further verified by real-time quantitative PCR (RT-qPCR) and methylation-specific PCR (MSP) in clinical samples. Furthermore, ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) was carried out on PCOS patients and healthy controls to identify differential lipid metabolites. Results Compared to the control group, 13,526 DMGs related to the promoter region and 2429 DEGs were found. The function analysis of DMGs and DEGs showed that they were mainly enriched in glycerophospholipid, ovarian steroidogenesis, and other lipid metabolic pathways. Moreover, 5753 genes in DMGs related to the promoter region were screened in the constructed PNA mice. Integrating the DMGs data from PCOS patients and PNA mice, we identified the following 8 genes: CDC42EP4, ERMN, EZR, PIK3R1, ARHGEF18, NECTIN2, TSC2, and TACSTD2. RT-qPCR and MSP verification results showed that the methylation and expression of TACSTD2 were consistent with sequencing data. Additionally, 15 differential lipid metabolites were shown in the serum of PCOS patients. The differential lipids were involved in glycerophospholipid and glycerolipid metabolism. Conclusion Using integration of methylome and lipid metabolites profiling we identified 8 potential epigenetic markers and 15 potential lipid metabolite markers for PCOS. Our results suggest that aberrant DNA methylation and lipid metabolite disorders may provide novel insights into the diagnosis and etiology of PCOS.
Collapse
Affiliation(s)
- Fei Zhang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yicen Ding
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Bohan Zhang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Mengju He
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Zhijiang Wang
- Department of Pharmaceutical Engineering, Zhejiang Pharmaceutical University, Ningbo, People’s Republic of China
| | - Chunbo Lu
- Department of Obstetrics and Gynecology, Qiuai Central Health Center, Ningbo, People’s Republic of China
| | - Yani Kang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
5
|
Nakanishi N, Osuka S, Kono T, Kobayashi H, Ikeda S, Bayasula B, Sonehara R, Murakami M, Yoshita S, Miyake N, Muraoka A, Kasahara Y, Murase T, Nakamura T, Goto M, Iwase A, Kajiyama H. Upregulated Ribosomal Pathway Impairs Follicle Development in a Polycystic Ovary Syndrome Mouse Model: Differential Gene Expression Analysis of Oocytes. Reprod Sci 2023; 30:1306-1315. [PMID: 36194357 DOI: 10.1007/s43032-022-01095-7] [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: 04/25/2022] [Accepted: 09/21/2022] [Indexed: 10/10/2022]
Abstract
Polycystic ovary syndrome (PCOS), a common endocrine disorder, is associated with impaired oocyte development, leading to infertility. However, the pathogenesis of PCOS has not been completely elucidated. This study aimed to determine the differentially expressed genes (DEGs) and epigenetic changes in the oocytes from a PCOS mouse model to identify the etiological factors. RNA-sequencing analysis revealed that 90 DEGs were upregulated and 27 DEGs were downregulated in mice with PCOS compared with control mice. DNA methylation analysis revealed 30 hypomethylated and 10 hypermethylated regions in the PCOS group. However, the DNA methylation status did not correlate with differential gene expression. The pathway enrichment analysis revealed that five DEGs (Rps21, Rpl36, Rpl36a, Rpl37a, and Rpl22l1) were enriched in ribosome-related pathways in the oocytes of mice with PCOS, and the immunohistochemical analysis revealed significantly upregulated expression levels of Rps21 and Rpl36. These results suggest that differential gene expression in the oocytes of mice in PCOS is related to impaired folliculogenesis. These findings improve our understanding of PCOS pathogenesis.
Collapse
Affiliation(s)
- Natsuki Nakanishi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
- Department of Maternal and Perinatal Medicine, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomohiro Kono
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Hisato Kobayashi
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Shinya Ikeda
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Bayasula Bayasula
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Reina Sonehara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Mayuko Murakami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sayako Yoshita
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Natsuki Miyake
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ayako Muraoka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yukiyo Kasahara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomohiko Murase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomoko Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Maki Goto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
6
|
Smirnov VV, Beeraka NM, Butko DY, Nikolenko VN, Bondarev SA, Achkasov EE, Sinelnikov MY, Vikram PRH. Updates on Molecular Targets and Epigenetic-Based Therapies for PCOS. Reprod Sci 2023; 30:772-786. [PMID: 35764857 DOI: 10.1007/s43032-022-01013-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022]
Abstract
Polycystic ovarian syndrome (PCOS) can cause infertility in females due to hyperandrogenism and neuroendocrine abnormalities. The aim of this study is to decipher the impact of endocrine variables, hyperandogenism, insulin resistance, oxidative stress, and dietary conditions in PCOS conditions, subsequently to depict the role of epigenetic factors relative to phenotypic manifestations in PCOS conditions. We have reviewed several metabolic milieus pertinent to PCOS conditions. Comparative efficacies of various PCOS therapies, and recent clinical recommendations for the effective management of PCOS and role of metabolic/endocrine variables in PCOS conditions were described. Comparative therapeutic effects were vividly delineated according to the variable pathophysiology and internal variables during PCOS syndrome on the female body through the formation of cascade of endocrine pathology, which affects working capacity and fosters redox stress-induced cardiovascular, neural, and liver abnormalities. GLP-1 agonists, insulin sensitizers (metformin), and diet and exercise regimens efficacy were explained in enhancing the fertility outcomes among the overweight or obese females with PCOS. Comprehensive appraisal of DNA methylation as epigenetic changes and the manifestations of methylated genes in PCOS conditions were discussed particularly to screen novel molecular targets for developing efficient diagnostic indicators for predicting PCOS risk or its progression. Due to the reversible nature of epigenetic modifications, it is possible to screen the "druggable" regions to target or to correct abnormalities in the gene expression subsequently to develop chromatin-modifying therapies against PCOS.
Collapse
Affiliation(s)
- Viktor V Smirnov
- Department of Medical Rehabilitation and Sports Medicine, Saint Petersburg State Pediatric Medical University of the Ministry of Health of the Russian Federation Saint Petersburg, Saint Petersburg, Russia
| | - Narasimha M Beeraka
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str, Moscow, 119991, Russia. .,Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR; A DST-FIST Supported Center), Department of Biochemistry, JSS Medical College, Mysuru, India.
| | - Dmitry Yu Butko
- Department of Medical Rehabilitation and Sports Medicine, Saint Petersburg State Pediatric Medical University of the Ministry of Health of the Russian Federation Saint Petersburg, Saint Petersburg, Russia
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str, Moscow, 119991, Russia.,Department of Normal and Topographic Anatomy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey A Bondarev
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str, Moscow, 119991, Russia
| | - Evgeniy E Achkasov
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str, Moscow, 119991, Russia
| | - P R Hemanth Vikram
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| |
Collapse
|
7
|
Rani S, Chandna P. Multiomics Analysis-Based Biomarkers in Diagnosis of Polycystic Ovary Syndrome. Reprod Sci 2023; 30:1-27. [PMID: 35084716 PMCID: PMC10010205 DOI: 10.1007/s43032-022-00863-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2023]
Abstract
Polycystic ovarian syndrome is an utmost communal endocrine, psychological, reproductive, and metabolic disorder that occurs in women of reproductive age with extensive range of clinical manifestations. This may even lead to long-term multiple morbidities including obesity, diabetes mellitus, insulin resistance, cardiovascular disease, infertility, cerebrovascular diseases, and ovarian and endometrial cancer. Women affliction from PCOS in midst assemblage of manifestations allied with menstrual dysfunction and androgen exorbitance, which considerably affects eminence of life. PCOS is recognized as a multifactorial disorder and systemic syndrome in first-degree family members; therefore, the etiology of PCOS syndrome has not been copiously interpreted. The disorder of PCOS comprehends numerous allied health conditions and has influenced various metabolic processes. Due to multifaceted pathophysiology engaging several pathways and proteins, single genetic diagnostic tests cannot be supportive to determine in straight way. Clarification of cellular and biochemical pathways and various genetic players underlying PCOS could upsurge our consideration of pathophysiology of this syndrome. It is requisite to know pathophysiological relationship between biomarker and their reflection towards PCOS disease. Biomarkers deliver vibrantly and potent ways to apprehend the spectrum of PCOS with applications in screening, diagnosis, characterization, and monitoring. This paper relies on the endeavor to point out many candidates as potential biomarkers based on omics technologies, thus highlighting correlation between PCOS disease with innovative technologies. Therefore, the objective of existing review is to encapsulate more findings towards cutting-edge advances in prospective use of biomarkers for PCOS disease. Discussed biomarkers may be fruitful in guiding therapies, addressing disease risk, and predicting clinical outcomes in future.
Collapse
Affiliation(s)
- Shikha Rani
- Department of Biophysics, University of Delhi, South Campus, Benito Juarez Road, New Delhi , 110021, India.
| | - Piyush Chandna
- Natdynamics Biosciences Confederation, Gurgaon, Haryana, 122001, India
| |
Collapse
|
8
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the Inflammatory Response in Polycystic Ovary Syndrome (PCOS)-Searching for Epigenetic Factors. Int J Mol Sci 2022; 23:ijms232314663. [PMID: 36498989 PMCID: PMC9736994 DOI: 10.3390/ijms232314663] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Despite its incidence, the syndrome is poorly understood and remains underdiagnosed, and female patients are diagnosed with a delay. The heterogenous nature of this complex disorder results from the combined occurrence of genetic, environmental, endocrine, and behavioral factors. Primary clinical manifestations of PCOS are derived from the excess of androgens (anovulation, polycystic ovary morphology, lack of or scanty, irregular menstrual periods, acne and hirsutism), whereas the secondary manifestations include multiple metabolic, cardiovascular, and psychological disorders. Dietary and lifestyle factors play important roles in the development and course of PCOS, which suggests strong epigenetic and environmental influences. Many studies have shown a strong association between PCOS and chronic, low-grade inflammation both in the ovarian tissue and throughout the body. In the vast majority of PCOS patients, elevated values of inflammatory markers or their gene markers have been reported. Development of the vicious cycle of the chronic inflammatory state in PCOS is additionally stimulated by hyperinsulinemia and obesity. Changes in DNA methylation, histone acetylation and noncoding RNA levels are presented in this review in the context of oxidative stress, reactive oxygen species, and inflammatory signaling in PCOS. Epigenetic modulation of androgenic activity in response to inflammatory signaling is also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
- Correspondence:
| | - Seweryn Trojanowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Anna Kociszewska
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
10
|
Rawat K, Sandhu A, Gautam V, Saha PK, Saha L. Role of genomic DNA methylation in PCOS pathogenesis: a systematic review and meta-analysis involving case controlled clinical studies. Mol Hum Reprod 2022; 28:6631266. [PMID: 35789386 DOI: 10.1093/molehr/gaac024] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
PCOS is often associated with aberrant DNA methylation. Despite the advances in diagnostics and treatment of PCOS, the pathophysiological mechanism remain unknown. Several genes are epigenetically dysregulated in PCOS and associated with pathological consequences of PCOS and metabolic comorbidities, however the methylation status of specific genes and to what extent the genes are deregulated in terms of methylation pattern are unknown. This review aimed to analyse the existing data for specific genes and find conclusive evidence of their involvement in PCOS and associated risks. A comprehensive literature search was conducted in five electronic databases. The case-controlled clinical studies using both PCOS and healthy women and evaluating the methylation pattern without any treatment or intervention were included in the study. A random-effect model was used to extract the data for meta-analysis, and outcomes were expressed as standardized mean difference with a 95% CI. From 541 screened records, 41 studies were included in the review and 21 of them were used for meta-analysis of 20 genes. Meta-analysis revealed a significant global DNA hypomethylation in different tissues and peripheral blood of patients with PCOS compared to healthy controls. Specific gene methylation assessment revealed that genes associated with several functions were significantly hypomethylated and hypermethylated in patients with PCOS. This review provides conclusive evidence of epigenetic deregulation of specific genes in PCOS. These genes can potentially be used to develop diagnostic biomarkers or as targets for personalised therapy.
Collapse
Affiliation(s)
- Kajal Rawat
- Department of Pharmacology Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, India- 160012
| | - Arushi Sandhu
- Department of Pharmacology Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, India- 160012
| | - Vipasha Gautam
- Department of Pharmacology Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, India- 160012
| | - Pradip Kumar Saha
- Department of Obstetrics and Gynecology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India- 160012
| | - Lekha Saha
- Department of Pharmacology Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, India- 160012
| |
Collapse
|
11
|
Scarfò G, Daniele S, Fusi J, Gesi M, Martini C, Franzoni F, Cela V, Artini PG. Metabolic and Molecular Mechanisms of Diet and Physical Exercise in the Management of Polycystic Ovarian Syndrome. Biomedicines 2022; 10:biomedicines10061305. [PMID: 35740328 PMCID: PMC9219791 DOI: 10.3390/biomedicines10061305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine systemic disorder mainly characterized by a hormonal and metabolic disbalance that leads to oligo/anovulation, hyperandrogenism and the formation of ovarian cysts. Despite the progress that has been reached in its diagnosis and management, little is known about the molecular mechanisms and signaling pathways underlying the pathogenic mechanisms. In this sense, recent research has suggested that the influence of multiple factors, including age, environment, lifestyle and the disease state environment can change the clinical presentation of PCOS via epigenetic modifications. Variants in the genes encoding for proteins involved in steroidogenesis and glucose homeostasis play a crucial role in the development of the disease. Other genes involved in inflammation and cell proliferation seem to undergo an epigenetic control. Moreover, lifestyle factors influence the PCOS course and prognosis, including diet and physical activity, which are fundamental in reducing oxidative stress, inflammation and in improving metabolic and hormonal parameters. In the present review, literature evidence on molecular and epigenetic mechanisms related to PCOS etiology will be discussed, with a particular attention on the positive influence of diet and physical activity as nonpharmacological ways of intervention in the management of the disease.
Collapse
Affiliation(s)
- Giorgia Scarfò
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.S.); (J.F.); (F.F.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Correspondence: (S.D.); (P.G.A.); Tel.: +39-050-2219608 (S.D.); +39-050-554104 (P.G.A.)
| | - Jonathan Fusi
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.S.); (J.F.); (F.F.)
| | - Marco Gesi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
| | - Ferdinando Franzoni
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.S.); (J.F.); (F.F.)
| | - Vito Cela
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Paolo Giovanni Artini
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
- Correspondence: (S.D.); (P.G.A.); Tel.: +39-050-2219608 (S.D.); +39-050-554104 (P.G.A.)
| |
Collapse
|
12
|
Guo Y, Peng X, Cheng R, Chen H, Luo X. Long non-coding RNA-X-inactive specific transcript inhibits cell viability, and induces apoptosis through the microRNA-30c-5p/Bcl2-like protein 11 signaling axis in human granulosa-like tumor cells. Bioengineered 2022; 13:14107-14117. [PMID: 35730492 PMCID: PMC9342309 DOI: 10.1080/21655979.2022.2080366] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The role of long noncoding RNAs (lncRNAs) is being actively explored in polycystic ovary syndrome (PCOS). Recent research has shown that long non-coding RNA (lncRNA) X–inactive Specific Transcript (XIST) is overexpressed in patients with PCOS and is associated with poor pregnancy outcomes. However, the precise function and mechanism of action of lncRNA XIST in PCOS are unknown. We aimed to determine whether lncRNA XIST contributes to PCOS by modulating ovarian granulosa cell physiology. We also investigated any potential molecular regulatory mechanisms. In this study, we discovered that the lncRNA XIST was significantly downregulated in human ovarian granulosa-like tumor (KGN) cells. Notably, overexpression of lncRNA XIST decreased miR-30c-5p expression in KGN cells, inhibited proliferation, and induced apoptosis in KGN cells. However, cotransfection with amiR-30c-5p mimic significantly reduced these effects. Additionally, we discovered that the miR-30c-5p mimic effectively inhibited Bcl2-like protein 11 (BCL2L11) expression, a critical apoptotic promoter, whereas silencing of miR-30c-5p increased BCL2L11 expression, inhibited KGN cell proliferation, and induced apoptosis. In contrast, cotransfection of BCL2L11 with siRNA significantly reversed these effects. In conclusion, this study established that lncRNA XIST plays a critical role in PCOS by modulating the miR-30c-5p/BCL2L11 signaling axis and regulating ovarian granulosa cell physiology.
Collapse
Affiliation(s)
- Yichuan Guo
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Xueping Peng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Ran Cheng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China.,Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China.,Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyan Luo
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China.,Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Sagvekar P, Shinde G, Mangoli V, Desai SK, Mukherjee S. Evidence for TET-mediated DNA demethylation as an epigenetic alteration in cumulus granulosa cells of women with polycystic ovary syndrome. Mol Hum Reprod 2022; 28:6595033. [PMID: 35640568 DOI: 10.1093/molehr/gaac019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral and tissue-specific alterations in global DNA methylation (5mC) and hydroxymethylation (5hmC) profiles have been charted as biomarkers for disease prediction and as hallmarks of dysregulated localized gene networks. Global and gene-specific epigenetic alterations in the 5mC profiles have shown widespread implications in etiology of polycystic ovary syndrome (PCOS). However, there has been no study in PCOS that integrates the quantification of 5mC and 5hmC signatures alongside the expression levels of DNA methylating and demethylating enzymes as respective indicators of methylation and demethylation pathways. Having previously shown that the 5mC signatures are not greatly altered in PCOS, we assessed the global 5hmC levels in peripheral blood leukocytes (PBLs) and cumulus granulosa cells (CGCs) of 40 controls and 40 women with PCOS. This analysis revealed higher 5hmC levels in CGCs of PCOS women, indicating a more dominant demethylation pathway. Further, we assessed the transcript and protein expression levels of DNA demethylating and methylating enzymes, i.e. ten-eleven translocation methylcytosine dioxygenases (TET1, TET2, TET3) and DNA methyltransferases (DNMT1, DNMT3A and DNMT3B), respectively, in CGCs. The relative transcript and protein expression levels of all three TETs were found to be higher in women with PCOS; and the TET mRNA expression profiles were positively correlated with 5hmC levels in CGCs. Also, all three DNMT genes showed altered transcript expression in PCOS, although only the downregulated DNMT3A transcript was correlated with decreasing 5mC levels. At the protein level, the expression of DNMT1 (maintenance methylation enzyme) was higher, while that of DNMT3A (denovo methylation enzyme) was found to be lower in PCOS compared to controls. Overall, these results indicate that DNA methylation changes in CGCs of PCOS women may arise partly due to intrinsic alterations in the transcriptional regulation of TETs and DNMT3A.
Collapse
Affiliation(s)
- Pooja Sagvekar
- Department of Molecular Endocrinology, National Institute for Research in Reproductive Health (ICMR-NIRRH), J.M. Street, Parel, Mumbai, 400012, India
| | - Gayatri Shinde
- Department of Molecular Endocrinology, National Institute for Research in Reproductive Health (ICMR-NIRRH), J.M. Street, Parel, Mumbai, 400012, India
| | - Vijay Mangoli
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai-, 400007, Maharashtra, India
| | - Sadhana K Desai
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai-, 400007, Maharashtra, India
| | - Srabani Mukherjee
- Department of Molecular Endocrinology, National Institute for Research in Reproductive Health (ICMR-NIRRH), J.M. Street, Parel, Mumbai, 400012, India
| |
Collapse
|
14
|
Divoux A, Erdos E, Whytock K, Osborne TF, Smith SR. Transcriptional and DNA Methylation Signatures of Subcutaneous Adipose Tissue and Adipose-Derived Stem Cells in PCOS Women. Cells 2022; 11:cells11050848. [PMID: 35269469 PMCID: PMC8909136 DOI: 10.3390/cells11050848] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is often associated with metabolic syndrome features, including central obesity, suggesting that adipose tissue (AT) is a key organ in PCOS pathobiology. In this study, we compared both abdominal (ABD) and gluteofemoral (GF) subcutaneous AT in women with and without PCOS. ABD and GF subcutaneous ATs from PCOS and BMI/WHR-matched control women were analyzed by RT-qPCR, FACS and histology. ABD and GF adipose-derived stem cell (ASC) transcriptome and methylome were analyzed by RNA-seq and DNA methylation array. Similar to the control group with abdominal obesity, the GF AT of PCOS women showed lower expression of genes involved in lipid accumulation and angiogenesis compared to ABD depot. FACS analysis revealed an increase in preadipocytes number in both AT depots from PCOS. Further pathway analysis of RNA-seq comparisons demonstrated that the ASCs derived from PCOS are pro-inflammatory and exhibit a hypoxic signature in the ABD depot and have lower expression of adipogenic genes in GF depot. We also found a higher CpG methylation level in PCOS compared to control exclusively in GF-ASCs. Our data suggest that ASCs play an important role in the etiology of PCOS, potentially by limiting expansion of the healthy lower-body AT.
Collapse
Affiliation(s)
- Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA; (K.W.); (S.R.S.)
- Correspondence: ; Tel.: +1-(407)-303-7100 (ext. 1101628)
| | - Edina Erdos
- Departments of Medicine and Biological Chemistry, Division of Diabetes Endocrinology and Metabolism, Institute for Fundamental Biomedical Research, Pediatrics Johns Hopkins University School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA; (E.E.); (T.F.O.)
| | - Katie Whytock
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA; (K.W.); (S.R.S.)
| | - Timothy F. Osborne
- Departments of Medicine and Biological Chemistry, Division of Diabetes Endocrinology and Metabolism, Institute for Fundamental Biomedical Research, Pediatrics Johns Hopkins University School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA; (E.E.); (T.F.O.)
| | - Steven R. Smith
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA; (K.W.); (S.R.S.)
| |
Collapse
|
15
|
Yao X, Li F, Wei Z, EI-Samahy MA, Feng X, Yang F, Wang F. Integrative Genome-Wide DNA Methylome and Transcriptome Analysis of Ovaries from Hu Sheep with High and Low Prolific. Front Cell Dev Biol 2022; 10:820558. [PMID: 35186931 PMCID: PMC8850840 DOI: 10.3389/fcell.2022.820558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/14/2022] [Indexed: 12/20/2022] Open
Abstract
DNA methylation plays an important role in biological processes by affecting gene expression. However, how DNA methylation regulates phenotypic variation in Hu sheep remains unclear. Therefore, we generated genome-wide DNA methylation and transcriptomic profiles in the ovaries of Hu sheep with different prolificacies and genotypes (FecBB and FecB+). Results showed that ovary DNA methylome and transcriptome were significantly different between high prolificacy and low prolificacy Hu sheep. Comparative methylome analyses identified 10,644, 9,594, and 12,214 differentially methylated regions and 87, 1,121, and 2,375 genes, respectively, showing differential expression levels in three different comparison groups. Female reproduction-associated differentially methylated regions-related genes and differentially expressed genes were enriched, thereby the respective interaction networks were constructed. Furthermore, systematical integrative analyses revealed a negative correlation between DNA methylation around the transcriptional start site and gene expression levels, which was confirmed by testing the expression of integrin β2 subunit (ITGB2) and lysosome-associated protein transmembrane-4 beta (LAPTM4B) in vivo and in vitro. These findings demonstrated that DNA methylation influences the propensity for prolificacy by affecting gene expression in the ovaries, which may contribute to a greater understanding of the epigenome and transcriptome that will be useful for animal breeding.
Collapse
Affiliation(s)
- Xiaolei Yao
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Fengzhe Li
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Zongyou Wei
- Taicang Agricultural and Rural Science and Technology Service Center, and Graduate Workstation, Taicang, China
| | - M. A. EI-Samahy
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
- Animal Production Research Institute, ARC, Ministry of Agriculture, Giza, Egypt
| | - Xu Feng
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Fan Yang
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Feng Wang
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Feng Wang,
| |
Collapse
|
16
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of polycystic ovary syndrome - challenges and opportunities for treatment. Nat Rev Endocrinol 2021; 17:521-533. [PMID: 34234312 DOI: 10.1038/s41574-021-00517-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and is associated with a substantially increased lifetime risk of comorbidities, including type 2 diabetes mellitus, psychiatric disorders and gynaecological cancers. Despite its high prevalence (~15%) and substantial economic burden, the aetiology of PCOS remains elusive. The genetic loci linked to PCOS so far account for only ~10% of its heritability, which is estimated at 70%. However, growing evidence suggests that altered epigenetic and developmental programming resulting from hormonal dysregulation of the maternal uterine environment contributes to the pathogenesis of PCOS. Male as well as female relatives of women with PCOS are also at an increased risk of developing PCOS-associated reproductive and metabolic disorders. Although PCOS phenotypes are highly heterogenous, hyperandrogenism is thought to be the principal driver of this condition. Current treatments for PCOS are suboptimal as they can only alleviate some of the symptoms; preventative and targeted treatments are sorely needed. This Review presents an overview of the current understanding of the aetiology of PCOS and focuses on the developmental origin and epigenetic inheritance of this syndrome.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Eiras MC, Pinheiro DP, Romcy KAM, Ferriani RA, Reis RMD, Furtado CLM. Polycystic Ovary Syndrome: the Epigenetics Behind the Disease. Reprod Sci 2021; 29:680-694. [PMID: 33826098 DOI: 10.1007/s43032-021-00516-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/21/2021] [Indexed: 10/21/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders, affecting approximately 5-20% of women of reproductive age. PCOS is a multifactorial, complex, and heterogeneous disease, characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovaries, which may lead to impaired fertility. Besides the reproductive outcomes, multiple comorbidities, such as metabolic disturbances, insulin resistance, obesity, diabetes, and cardiovascular disease, are associated with PCOS. In addition to the clear genetic basis, epigenetic alterations may also play a central role in PCOS outcomes, as environmental and hormonal alterations directly affect clinical manifestations and PCOS development. Here, we highlighted the epigenetic modifications in the multiplicity of clinical manifestations, as well as environmental epigenetic disruptors, as intrauterine hormonal and metabolic alterations affecting embryo development and the adulthood lifestyle, which may contribute to PCOS development. Additionally, we also discussed the new approaches for future studies and potential epigenetic biomarkers for the treatment of associated comorbidities and improvement in quality of life of women with PCOS.
Collapse
Affiliation(s)
- Matheus Credendio Eiras
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of Sao Paulo, Av Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, SP, Brazil
| | - Daniel Pascoalino Pinheiro
- Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceara, Coronel Nunes de Melo, 1000, Rodolfo Teófilo, Fortaleza, 60430-275, CE, Brazil
| | - Kalil Andrade Mubarac Romcy
- Drug Research and Development Center, Postgraduate Program in Medical and Surgical Sciences, Federal University of Ceara, Coronel Nunes de Melo, 1000, Rodolfo Teófilo, Fortaleza, CE, 60430-275, Brazil
| | - Rui Alberto Ferriani
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of Sao Paulo, Av Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, SP, Brazil
| | - Rosana Maria Dos Reis
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of Sao Paulo, Av Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, SP, Brazil.
| | - Cristiana Libardi Miranda Furtado
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of Sao Paulo, Av Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, SP, Brazil. .,Drug Research and Development Center, Postgraduate Program in Medical and Surgical Sciences, Federal University of Ceara, Coronel Nunes de Melo, 1000, Rodolfo Teófilo, Fortaleza, CE, 60430-275, Brazil.
| |
Collapse
|
18
|
Barnard L, du Toit T, Swart AC. Back where it belongs: 11β-hydroxyandrostenedione compels the re-assessment of C11-oxy androgens in steroidogenesis. Mol Cell Endocrinol 2021; 525:111189. [PMID: 33539964 DOI: 10.1016/j.mce.2021.111189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/29/2022]
Abstract
Adrenal steroidogenesis has, for decades, been depicted as three biosynthesis pathways -the mineralocorticoid, glucocorticoid and androgen pathways with aldosterone, cortisol and androstenedione as the respective end products. 11β-hydroxyandrostenedione was not included as an adrenal steroid despite the adrenal output of this steroid being twice that of androstenedione. While it is the end of the line for aldosterone and cortisol, as it is in these forms that they exhibit their most potent receptor activities prior to inactivation and conjugation, 11β-hydroxyandrostenedione is another matter entirely. The steroid, which is weakly androgenic, has its own designated pathway yielding 11-ketoandrostenedione, 11β-hydroxytestosterone and the potent androgens, 11-ketotestosterone and 11-ketodihydrotestosterone, primarily in the periphery. Over the last decade, these C11-oxy C19 steroids have once again come to the fore with the rising number of studies contradicting the generally accepted notion that testosterone and it's 5α-reduced product, dihydrotestosterone, are the principal potent androgens in humans. These C11-oxy androgens have been shown to contribute to the androgen milieu in adrenal disorders associated with androgen excess and in androgen dependant disease progression. In this review, we will highlight these overlooked C11-oxy C19 steroids as well as the C11-oxy C21 steroids and their contribution to congenital adrenal hyperplasia, polycystic ovarian syndrome and prostate cancer. The focus is on new findings over the past decade which are slowly but surely reshaping our current outlook on human sex steroid biology.
Collapse
Affiliation(s)
- Lise Barnard
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Therina du Toit
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Amanda C Swart
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa; Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa.
| |
Collapse
|
19
|
Almalki WH. A review on inositol's potential in cyclic disturbances of adipose-endocrinology-associated polycystic ovary syndrome. Mol Cell Biochem 2021; 476:2943-2949. [PMID: 33755878 DOI: 10.1007/s11010-021-04123-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/25/2021] [Indexed: 10/21/2022]
Abstract
Since the lack of certainty in identifying polycystic ovary syndrome (PCOS) demonstrates confusion regarding the disorder's pathophysiology and its therapeutic approaches, systematic screening of women under diagnostic guidelines of the NIH reported that about 4-10 percent of reproductive women aged 20-44 years suffer from PCOS. Not all females with PCOS-defining biochemical and clinical characteristics and about 22% of PCOS women have no symptoms. PCOS is a heterogeneous phenotypic and clinical condition, combined with metabolic implications. The root cause of PCOS is the major issue of IR or irregular androgen secretion and constant effort is being made in identifying the dynamic pathogenic network underlying the syndrome. Regardless of PCOS initiating cause, IR therapy and hyperinsulinemia can restore metabolic and hormonal homeostasis, and minimize ovarian dysfunction. Thus, the impact of insulin on ovaries in hyperinsulinemic individuals can account for many of the PCOS characteristics and is important for developing treatment strategies. Therefore, our primary aim is to investigate the proper understanding of endocrine disruption during PCOS and secondary to the therapeutic potential of inositol in reestablishing the equilibrium of ovarian dysfunction, anovulation, and eventually infertility.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| |
Collapse
|
20
|
Yang D, Wang Y, Zheng Y, Dai F, Liu S, Yuan M, Deng Z, Bao A, Cheng Y. Silencing of lncRNA UCA1 inhibited the pathological progression in PCOS mice through the regulation of PI3K/AKT signaling pathway. J Ovarian Res 2021; 14:48. [PMID: 33743811 PMCID: PMC7980617 DOI: 10.1186/s13048-021-00792-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is the most common hormonal disorder among reproductive-aged women worldwide, however, the mechanisms and progression of PCOS still unclear due to its heterogeneous nature. Using the human granulosa-like tumor cell line (KGN) and PCOS mice model, we explored the function of lncRNA UCA1 in the pathological progression of PCOS. Results CCK8 assay and Flow cytometry were used to do the cell cycle, apoptosis and proliferation analysis, the results showed that UCA1 knockdown in KGN cells inhibited cell proliferation by blocking cell cycle progression and promoted cell apoptosis. In the in vivo experiment, the ovary of PCOS mice was injected with lentivirus carrying sh-UCA1, the results showed that knockdown of lncRNA UCA1 attenuated the ovary structural damage, increased the number of granular cells, inhibited serum insulin and testosterone release, and reduced the pro-inflammatory cytokine production. Western blot also revealed that UCA1 knockdown in PCOS mice repressed AKT activation, inhibitor experiment demonstrated that suppression of AKT signaling pathway, inhibited the cell proliferation and promoted apoptosis. Conclusions Our study revealed that, in vitro, UCA1 knockdown influenced the apoptosis and proliferation of KGN cells, in vivo, silencing of UCA1 regulated the ovary structural damage, serum insulin release, pro-inflammatory production, and AKT signaling pathway activation, suggesting lncRNA UCA1 plays an important role in the pathological progression of PCOS.
Collapse
Affiliation(s)
- Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Anyu Bao
- Department of Clinical Laboratoy, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Mimouni NEH, Paiva I, Barbotin AL, Timzoura FE, Plassard D, Le Gras S, Ternier G, Pigny P, Catteau-Jonard S, Simon V, Prevot V, Boutillier AL, Giacobini P. Polycystic ovary syndrome is transmitted via a transgenerational epigenetic process. Cell Metab 2021; 33:513-530.e8. [PMID: 33539777 PMCID: PMC7928942 DOI: 10.1016/j.cmet.2021.01.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/20/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common reproductive and metabolic disorder affecting women of reproductive age. PCOS has a strong heritable component, but its pathogenesis has been unclear. Here, we performed RNA sequencing and genome-wide DNA methylation profiling of ovarian tissue from control and third-generation PCOS-like mice. We found that DNA hypomethylation regulates key genes associated with PCOS and that several of the differentially methylated genes are also altered in blood samples from women with PCOS compared with healthy controls. Based on this insight, we treated the PCOS mouse model with the methyl group donor S-adenosylmethionine and found that it corrected their transcriptomic, neuroendocrine, and metabolic defects. These findings show that the transmission of PCOS traits to future generations occurs via an altered landscape of DNA methylation and propose methylome markers as a possible diagnostic landmark for the condition, while also identifying potential candidates for epigenetic-based therapy.
Collapse
Affiliation(s)
- Nour El Houda Mimouni
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France
| | - Isabel Paiva
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), 12 Rue Goethe, Strasbourg 67000, France
| | - Anne-Laure Barbotin
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France
| | - Fatima Ezzahra Timzoura
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Stephanie Le Gras
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France
| | - Pascal Pigny
- CHU Lille, Service de Biochimie et Hormonologie, Centre de Biologie Pathologie, Lille, France
| | - Sophie Catteau-Jonard
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France; CHU Lille, Service de Gynécologie Médicale, Hôpital Jeanne de Flandre, Lille, France
| | - Virginie Simon
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France; CHU Lille, Service de Gynécologie Médicale, Hôpital Jeanne de Flandre, Lille, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France
| | - Anne-Laurence Boutillier
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), 12 Rue Goethe, Strasbourg 67000, France.
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 days for health, 59000 Lille, France.
| |
Collapse
|
22
|
Abstract
PCOS is a common and heterogeneous endocrine disorder in women of reproductive age, frequently associated with metabolic abnormalities. It was estimated that about 75% of these subjects have an impairment of insulin action, as measured by gold standard methods. While the relationship between insulin resistance and PCOS is consistently shown by a number of studies, the mechanisms underlying its primary origin still remains an unsolved issue. Insulin resistance and the associated hyperinsulinemia can induce both the endocrine and reproductive traits of PCOS. However, androgen excess, in turn, can impair insulin action, directly and/or through several changes occurring in different tissues. Body fat excess, which is another common feature in these women, can contribute to worsening the whole picture. Nevertheless, insulin resistance may also be found in many normal-weight individuals. Endocrine and metabolic abnormalities can develop in different moments, and probably there is fetal programming of these alterations. However, a number of vicious circles, with bidirectional relationships between androgen excess and insulin resistance, and with the contribution of several other factors, make it extremely difficult to understand where this process really originates. This review summarizes available evidence on this topic, in order to better understand the complex relationships linking hyperandrogenism and impaired insulin action in women with PCOS.
Collapse
Affiliation(s)
- P Moghetti
- Unit of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, P.le Stefani, 1, 37126, Verona, Italy.
| | - F Tosi
- Unit of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, P.le Stefani, 1, 37126, Verona, Italy
| |
Collapse
|
23
|
Motta AB. Epigenetic Marks in Polycystic Ovary Syndrome. Curr Med Chem 2021; 27:6727-6743. [PMID: 31580245 DOI: 10.2174/0929867326666191003154548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022]
Abstract
Polycystic Ovary Syndrome (PCOS) is a common endocrine and metabolic disorder that affects women in their reproductive age. Recent studies have shown that genes have an important role in the etiology of PCOS. However, the precise way in which these genes are transcriptionally and post-transcriptionally regulated is poorly understood. The aim of the present review is to provide updated information on miRNAs and DNA methylation as epigenetic marks of PCOS. The data presented here allow concluding that both microRNAs and DNA methylation can be considered as possible useful biomarkers when choosing the treatment for a specific PCOS phenotype and thus represent two important tools for the diagnosis and treatment of PCOS patients.
Collapse
Affiliation(s)
- Alicia Beatriz Motta
- Laboratorio de Fisio-patologia Ovarica, Centro de Estudios Farmacologicos y Botanicos (CEFYBO), Consejo Nacional de Investigaciones Cientificas y Tecnologicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Paraguay 2155, CP1121, Ciudad Autonoma de Buenos Aires, Argentina
| |
Collapse
|
24
|
Molecular Targets and Associated Signaling Pathways of Jingshu Granules in Ovarian Cysts Based on Systemic Pharmacological Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6660087. [PMID: 33623786 PMCID: PMC7875638 DOI: 10.1155/2021/6660087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/09/2021] [Indexed: 11/18/2022]
Abstract
Background More than a third of women could develop ovarian cysts during their lifetime. Jingshu granules are used for the treatment of gynecological disease of primary dysmenorrhea. However, the molecular mechanisms of Jingshu granules in ovarian cysts are still unreported. We aimed to find the active ingredients, molecular targets, and potential signaling pathways of Jingshu granules in ovarian cysts by using the systemic pharmacological analysis. Methods Firstly, the effect of Jingshu granules on female hormones and reproductive organs of young female rats was evaluated. Secondly, candidate pharmaceutical ingredients of Jingshu granules were retrieved from the traditional Chinese medicine systems pharmacology (TCMSP) database and analysis platform. Potential protein targets for the active ingredients in Jingshu granules were then identified according to the oral bioavailability and drug-likeness indices. Thirdly, ovarian cyst-related gene targets were screened based on different databases. Finally, enrichment analysis was used to analyze the potential biological function of intersection targets between Jingshu granules and ovarian cysts. Results In young female rats, Jingshu granules reduced the secretion of estradiol, progesterone, and prolactin and could affect the development of the uterus. This suggested that Jingshu granules played roles in hormone secretion and reproduction. From the TCMSP, a total of 1021 pharmaceutical ingredients of Jingshu granules were retrieved. After further screening, a total of 166 active ingredients and 159 protein targets of Jingshu granules were identified. In addition, 4488 gene targets of ovarian cysts were screened out. After taking the intersection, a total of 110 intersection targets were identified between potential protein targets of Jingshu granules and gene targets of ovarian cysts. In the functional analysis of 110 intersection targets, 8 signaling pathways including progesterone-mediated oocyte maturation (MAPK8 and CDK1 involved), GnRH signaling pathway (JUN involved), T cell receptor signaling pathway and Toll-like receptor signaling pathway (MAPK1 involved), NOD-like receptor signaling pathway (TNF, IL6, and IL1B involved), p53 signaling pathway (CDK2 and CDK4 involved), VEGF signaling pathway (MAPK14 involved), and PPAR signaling pathway (PPARG involved) were obtained. Conclusion Our study revealed that Jingshu granules could function in patients with ovarian cysts through a number of molecular targets and signaling pathways. Our study may provide a new field into the mechanisms of Jingshu granules in ovarian cysts, from the molecular to the signaling pathway level.
Collapse
|
25
|
Ghasemi M, Heidari Nia M, Hashemi M, Keikha N, Fazeli K, Taji O, Naghavi A. An association study of polymorphisms in the H19 imprinted gene in an Iranian population with the risk of polycystic ovary syndrome. Biol Reprod 2020; 103:978-985. [PMID: 32720692 DOI: 10.1093/biolre/ioaa131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/12/2020] [Accepted: 07/22/2020] [Indexed: 11/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies that causes problems in female fertility at the reproductive age. PCOS is a multifactorial disease, with genetic factors playing a crucial role in its development. H19 is a long non-coding RNA (lncRNA) expressed from the maternal chromosome, which is correlated with PCOS. In this study, 115 women suffering from PCOS and 130 healthy women with regular menstrual cycles were recruited as case and control groups, respectively. After the extraction of genomic DNA, the restriction fragment length polymorphism polymerase chain reaction was employed for genotyping of rs2067051G>A and rs3741219T>C. Statistical analysis was done using SPSS package V.22 for Windows. In silico analysis was recruited to determine the effects of SNPs on the secondary structure of gene transcript as well as miRNA binding sites. The obtained data showed that the A allele of rs2067051G>A was associated with the high risk of PCOS (OR = 2.00, 95%CI = 1.38-2.91, P = 0.00). AG and AA genotypes led to a 3.64- and (about) a five-fold increase in the risk of PCOS, respectively (95%CI = 2.02-6.54, P = 0.00, and 95%CI = 1.51-16.52, P = 0.00, respectively). These variants caused a significant increase in the risk of this disorder in all genotype models except in the recessive model. However, no association was found between rs3741219T>C and the increased risk of PCOS, either in the allele or in the genotype models. According to the findings, rs2067051G>A is associated with an increased risk of PCOS in the Iranian population.
Collapse
Affiliation(s)
- Marzieh Ghasemi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan, Iran.,Moloud Infertility Center, Ali-ibn-Abitaleb Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Narjes Keikha
- Moloud Infertility Center, Ali-ibn-Abitaleb Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Kimia Fazeli
- School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Omid Taji
- Medical Genetic Reference Laboratory, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Anoosh Naghavi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Genetics, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
26
|
Sun Y, Wu Q, Pan J, Li T, Liu L, Chen D, Zhang X, Chen H, Li Y, Lin R. Identification of differentially expressed genes and signalling pathways in the ovary of higher and lower laying ducks. Br Poult Sci 2020; 61:609-614. [PMID: 33012177 DOI: 10.1080/00071668.2020.1792834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
1. Ovarian transcriptomic profiling between birds showing high egg number (HEN) and birds of low egg number (LEN) in Longyan Shan-ma ducks at 71 weeks of age was carried out using Illumina Hiseq 2500 technology. 2. A total of 343 differentially expressed genes (DEGs, 269 upregulated and 74 downregulated) were identified between HEN and LEN ovaries. These DEGs were enriched in 30 Gene Ontology terms. Pathway functional analysis found that the DEGs were enriched in 10 metabolic pathways (P < 0.05), one of which was regulation of the actin cytoskeleton pathway (Q < 0.05). 3. Three integrin family genes, ITGB2, ITGB5 and ITGA8 were differentially expressed in the RNA-seq and qPCR experiments. 4. The DEGs and signalling pathways identified in ovarian tissue in this study provide new insights into high egg production in Longyan Shan-ma duck.
Collapse
Affiliation(s)
- Y Sun
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China.,Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology , Longyan, Fujian, P.R. China.,Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Fujian Province University , Longyan, Fujian, P.R. China
| | - Q Wu
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China.,Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology , Longyan, Fujian, P.R. China.,Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Fujian Province University , Longyan, Fujian, P.R. China
| | - J Pan
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China
| | - T Li
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China
| | - L Liu
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China
| | - D Chen
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China
| | - X Zhang
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China
| | - H Chen
- Longyan Shan-ma Duck Original Breeding Farm, Agricultural Bureau of Xinluo District , Longyan, P.R. China
| | - Y Li
- College of Life Science, Longyan University , Longyan, Fujian, P.R. China.,Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology , Longyan, Fujian, P.R. China.,Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), Fujian Province University , Longyan, Fujian, P.R. China
| | - R Lin
- Longyan Shan-ma Duck Original Breeding Farm, Agricultural Bureau of Xinluo District , Longyan, P.R. China
| |
Collapse
|
27
|
Butler AE, Hayat S, Dargham SR, Malek JA, Abdullah SA, Mahmoud YA, Sathyapalan T, Atkin SL. Long non-coding RNA expression in non-obese women with polycystic ovary syndrome and weight-matched controls. Reprod Biomed Online 2020; 41:579-583. [PMID: 32819839 DOI: 10.1016/j.rbmo.2020.06.018] [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: 05/19/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022]
Abstract
RESEARCH QUESTION Long non-coding RNA (lncRNA) do not show protein translation but do have gene regulatory functions in several disease states. Studies have shown that lncRNA differ in overweight women with polycystic ovary syndrome (PCOS), increased insulin resistance and hyperandrogenaemia. The objective of this study was to determine the lncRNA in serum in age- and weight-matched non-obese women with and without PCOS. METHODS In this prospective pilot cohort study, lncRNA were measured in serum in 13 non-obese women with PCOS and 10 control women undergoing IVF. RESULTS There was no difference between groups in terms of age, body mass index or insulin resistance. Women with PCOS showed a higher free androgen index (FAI; P = 0.03) and anti-Müllerian hormone (AMH) concentration (P = 0.001). A total of 29 lncRNA (P ≤ 0.05) differed between PCOS groups. lncRNA AC095350.1 correlated with age (r = 0.79, P = 0.04), but no correlation was seen between the significantly different lncRNA and FAI or AMH values. Functional pathway assessment using the Ingenuity Pathway Assessment tool showed no relationships for the lncRNA. CONCLUSION lncRNA in serum differed between non-obese women with PCOS and the control group, and the pattern of expression differed from that reported in obese women with PCOS from the same ethnic population; however, it but did not correlate with androgen or insulin resistance.
Collapse
Affiliation(s)
- Alexandra E Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| | | | | | - Joel A Malek
- Weill Cornell Medical College in Qatar, Doha, Qatar
| | | | | | - Thozhukat Sathyapalan
- Academic Diabetes and Endocrinology, Hull York Medical School, University of Hull, Hull, UK
| | - Stephen L Atkin
- Royal College of Surgeons in Ireland-Bahrain, Busaiteen, Bahrain
| |
Collapse
|
28
|
Liu L, He D, Wang Y, Sheng M. Integrated analysis of DNA methylation and transcriptome profiling of polycystic ovary syndrome. Mol Med Rep 2020; 21:2138-2150. [PMID: 32323770 PMCID: PMC7115196 DOI: 10.3892/mmr.2020.11005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to identify potentially important biomarkers associated with polycystic ovary syndrome (PCOS) by integrating DNA methylation with transcriptome profiling. The transcription (E‑MTAB‑3768) and methylation (E‑MTAB‑3777) datasets were retrieved from ArrayExpress. Paired transcription and methylation profiling data of 10 cases of PCOS and 10 healthy controls were available for screening differentially expressed genes (DEGs) and differentially methylated genes (DMGs). Genes with a negative correlation between expression levels and methylation levels were retained by correlation analysis to construct a protein‑protein interaction (PPI) network. Subsequently, functional and pathway enrichment analyses were performed to identify genes in the PPI network. Additionally, a disease‑associated pathway network was also established. A total of 491 overlapping genes, and the expression levels of 237 genes, were negatively correlated with their methylation levels. Functional enrichment analysis revealed that genes in the PPI network were mainly involved with biological processes of cellular response to stress, negative regulation of the biosynthetic process, and regulation of cell proliferation. The constructed pathway network associated with PCOS led to the identification of four important genes (SPP1, F2R, IL12B and RBP4) and two important pathways (Jak‑STAT signaling pathway and neuroactive ligand‑receptor interaction). Taken, together, the results from the present study have revealed numerous important genes with abnormal DNA methylation levels and altered mRNA expression levels, along with their associated functions and pathways. These findings may contribute to an improved understanding of the possible pathophysiology of PCOS.
Collapse
Affiliation(s)
- Li Liu
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Dongyun He
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Yang Wang
- Department of Dermatology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130031, P.R. China
| | - Minjia Sheng
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| |
Collapse
|
29
|
Echiburú B, Milagro F, Crisosto N, Pérez-Bravo F, Flores C, Arpón A, Salas-Pérez F, Recabarren SE, Sir-Petermann T, Maliqueo M. DNA methylation in promoter regions of genes involved in the reproductive and metabolic function of children born to women with PCOS. Epigenetics 2020; 15:1178-1194. [PMID: 32283997 DOI: 10.1080/15592294.2020.1754674] [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] [Indexed: 12/28/2022] Open
Abstract
Clinical and experimental evidences indicate that epigenetic modifications induced by the prenatal environment are related to metabolic and reproductive derangements in polycystic ovary syndrome (PCOS). Alterations in the leptin and adiponectin systems, androgen signalling and antimüllerian hormone (AMH) levels have been observed in PCOS women and in their offspring. Using a targeted Next-Generation Sequencing (NGS), we studied DNA methylation in promoter regions of the leptin (LEP), leptin receptor (LEPR), adiponectin (ADIPOQ), adiponectin receptor 1 and 2 (ADIPOR1 and ADIPOR2), AMH and androgen receptor (AR) genes in 24 sons and daughters of women with PCOS (12 treated with metformin during pregnancy) and 24 children born to non-PCOS women during early infancy (2-3 months of age). Genomic DNA was extracted from whole blood, bisulphite converted and sequenced by NGS. Girls showed differences between groups in 1 CpG site of LEPR, 2 of LEP, 1 of ADIPOR2 and 2 of AR. Boys showed differences in 5 CpG sites of LEP, 3 of AMH and 9 of AR. Maternal metformin treatment prevented some of these changes in LEP, ADIPOR2 and partially in AR in girls, and in LEP and AMH in boys. Maternal BMI at early pregnancy was inversely correlated with the methylation levels of the ChrX-67544981 site in the whole group of girls (r = -0.530, p = 0.008) and with the global Z-score in all boys (r = -0.539, p = 0.007). These data indicate that the intrauterine PCOS environment predisposes the offspring to acquire certain sex-dependent DNA methylation patterns in the promoter regions of metabolic and reproductive genes.
Collapse
Affiliation(s)
- Bárbara Echiburú
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Fermín Milagro
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain.,Centro De Investigación Biomédica En Red Fisiopatología De La Obesidad Y Nutrición (Ciberobn), Instituto De Salud Carlos III , Madrid, Spain
| | - Nicolás Crisosto
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile.,Unit of Endocrinology, Clínica Las , Santiago, Chile
| | - Francisco Pérez-Bravo
- Laboratory of Nutritional Genomics, Department of Nutrition, Faculty of Medicine, University of Chile , Santiago, Chile
| | - Cristian Flores
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Ana Arpón
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain
| | - Francisca Salas-Pérez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain
| | - Sergio E Recabarren
- Laboratory of Animal Physiology and Endocrinology, Department of Animal Science, Faculty of Veterinary Sciences, University of Concepcion , Chillán, Chile
| | - Teresa Sir-Petermann
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Manuel Maliqueo
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| |
Collapse
|
30
|
|
31
|
Makrinou E, Drong AW, Christopoulos G, Lerner A, Chapa-Chorda I, Karaderi T, Lavery S, Hardy K, Lindgren CM, Franks S. Genome-wide methylation profiling in granulosa lutein cells of women with polycystic ovary syndrome (PCOS). Mol Cell Endocrinol 2020; 500:110611. [PMID: 31600550 PMCID: PMC7116598 DOI: 10.1016/j.mce.2019.110611] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/20/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023]
Abstract
Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder amongst women of reproductive age, whose aetiology remains unclear. To improve our understanding of the molecular mechanisms underlying the disease, we conducted a genome-wide DNA methylation profiling in granulosa lutein cells collected from 16 women suffering from PCOS, in comparison to 16 healthy controls. Samples were collected by follicular aspiration during routine egg collection for IVF treatment. Study groups were matched for age and BMI, did not suffer from other disease and were not taking confounding medication. Comparing women with polycystic versus normal ovarian morphology, after correcting for multiple comparisons, we identified 106 differentially methylated CpG sites with p-values <5.8 × 10-8 that were associated with 88 genes, several of which are known to relate either to PCOS or to ovarian function. Replication and validation of the experiment was done using pyrosequencing to analyse six of the identified differentially methylated sites. Pathway analysis indicated potential disruption in canonical pathways and gene networks that are, amongst other, associated with cancer, cardiogenesis, Hedgehog signalling and immune response. In conclusion, these novel findings indicate that women with PCOS display epigenetic changes in ovarian granulosa cells that may be associated with the heterogeneity of the disorder.
Collapse
Affiliation(s)
- E Makrinou
- Imperial College London, Faculty of Medicine, Institute of Reproductive and Developmental Biology, London, W12 0NN, UK.
| | - A W Drong
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, OX3 7LF, UK
| | - G Christopoulos
- IVF Unit, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0NN, UK
| | - A Lerner
- Imperial College London, Faculty of Medicine, Institute of Reproductive and Developmental Biology, London, W12 0NN, UK
| | - I Chapa-Chorda
- Imperial College London, Faculty of Medicine, Institute of Reproductive and Developmental Biology, London, W12 0NN, UK
| | - T Karaderi
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus
| | - S Lavery
- IVF Unit, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0NN, UK
| | - K Hardy
- Imperial College London, Faculty of Medicine, Institute of Reproductive and Developmental Biology, London, W12 0NN, UK
| | - C M Lindgren
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, OX3 7LF, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - S Franks
- Imperial College London, Faculty of Medicine, Institute of Reproductive and Developmental Biology, London, W12 0NN, UK
| |
Collapse
|
32
|
Butler AE, Ramachandran V, Cunningham TK, David R, Gooderham NJ, Benurwar M, Dargham SR, Hayat S, Sathyapalan T, Najafi-Shoushtari SH, Atkin SL. Increased MicroRNA Levels in Women With Polycystic Ovarian Syndrome but Without Insulin Resistance: A Pilot Prospective Study. Front Endocrinol (Lausanne) 2020; 11:571357. [PMID: 33101204 PMCID: PMC7556216 DOI: 10.3389/fendo.2020.571357] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Small noncoding microRNA (miRNA) have regulatory functions in polycystic ovary syndrome (PCOS) that differ to those in women without PCOS. However, little is known about miRNA expression in women with PCOS who are not insulin resistant (IR). METHODS Circulating miRNAs were measured using quantitative polymerase chain reaction (qPCR) in 24 non-obese BMI and age matched women with PCOS and 24 control women. A miRNA data set was used to determine miRNA levels. RESULTS Women with PCOS showed a higher free androgen index (FAI) and anti-mullerian hormone (AMH) but IR did not differ. Four miRNAs (miR-1260a, miR-18b-5p, miR-424-5p, and miR let-7b-3p) differed between control and PCOS women that passed the false discovery rate (FDR) out of a total of 177 circulating miRNAs that were detected. MiRNA let-7b-3p correlated with AMH in PCOS (p < 0.05). When the groups were combined, miR-1260a correlated with FAI and let-7b-3p correlated with body mass index (BMI) (p < 0.05). There was no correlation to androgen levels. Ingenuity pathway analysis showed that nine of the top 10 miRNAs reported were associated with inflammatory pathways. CONCLUSION When IR did not differ between PCOS and control women, only four miRNA differed significantly suggesting that IR may be a driver for many of the miRNA changes reported. Let-7b-3p was related to AMH in PCOS, and to BMI as a group, whilst miR-1260a correlated with FAI. Androgen levels, however, had no effect upon circulating miRNA profiles. The expressed miRNAs were associated with the inflammatory pathway involving TNF and IL6.
Collapse
Affiliation(s)
- Alexandra E. Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- *Correspondence: Alexandra E. Butler, ;
| | - Vimal Ramachandran
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Thomas Keith Cunningham
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Heslington, United Kingdom
| | - Rhiannon David
- Department of Surgery & Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Nigel J. Gooderham
- Department of Surgery & Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Manasi Benurwar
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Soha R. Dargham
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Shahina Hayat
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Heslington, United Kingdom
| | - S Hani Najafi-Shoushtari
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, United States
| | - Stephen L. Atkin
- Postgraduate Studies and Research, Royal College of Surgeons Ireland, Al Muharraq, Bahrain
| |
Collapse
|
33
|
Wen L, Liu Q, Xu J, Liu X, Shi C, Yang Z, Zhang Y, Xu H, Liu J, Yang H, Huang H, Qiao J, Tang F, Chen ZJ. Recent advances in mammalian reproductive biology. SCIENCE CHINA. LIFE SCIENCES 2020; 63:18-58. [PMID: 31813094 DOI: 10.1007/s11427-019-1572-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/22/2019] [Indexed: 01/05/2023]
Abstract
Reproductive biology is a uniquely important topic since it is about germ cells, which are central for transmitting genetic information from generation to generation. In this review, we discuss recent advances in mammalian germ cell development, including preimplantation development, fetal germ cell development and postnatal development of oocytes and sperm. We also discuss the etiologies of female and male infertility and describe the emerging technologies for studying reproductive biology such as gene editing and single-cell technologies.
Collapse
Affiliation(s)
- Lu Wen
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qiang Liu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Jingjing Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Xixi Liu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Chaoyi Shi
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Zuwei Yang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yili Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Hong Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Jiang Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Hui Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Hefeng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Jie Qiao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Zi-Jiang Chen
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250021, China.
| |
Collapse
|
34
|
Butler AE, Ramachandran V, Sathyapalan T, David R, Gooderham NJ, Benurwar M, Dargham SR, Hayat S, Hani Najafi-Shoushtari S, Atkin SL. microRNA Expression in Women With and Without Polycystic Ovarian Syndrome Matched for Body Mass Index. Front Endocrinol (Lausanne) 2020; 11:206. [PMID: 32411089 PMCID: PMC7199502 DOI: 10.3389/fendo.2020.00206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 03/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Despite several authors who have hypothesized that alterations of small noncoding RNAs (miR) are implicated in the etiopathogenesis of polycystic ovarian syndrome (PCOS), contrasting findings have been reported so far. Discrepancies in body mass index (BMI) levels may account for these differences; therefore, the aim of the present study was to determine whether miR differed in serum samples collected from age- and BMI-matched control and PCOS women. Methods: In a cross-sectional study, miR were measured using quantitative polymerase chain reaction in 29 women with anovulatory PCOS women and 29 control women who were in the follicular phase of their menstrual cycle, from the local biobank. Results: One hundred seventy-six miR were detected, of which 15 miR passed the false discovery rate (FDR; p < 0.05) that differed between PCOS and control women. There was no association of the top 9 miR (p < 0.02) (miR-486-5p, miR-24-3p, miR-19b-3p, miR-22-3p, miR-19a-3p, miR-339-5p, miR-185-5p, miR-101-3p, miR-let-7i-5p) with BMI, androgen levels, insulin resistance, or antimullerian hormone (AMH) in either PCOS or normal women. Ingenuity pathway assessment showed the pathways were interrelated for abnormalities of the reproductive system. Conclusion: When the confounding influence of weight was accounted for, miR levels differed between anovulatory PCOS women and control women in the follicular phase of the menstrual cycle. Interestingly, the differing miR were associated with the pathways of reproductive abnormalities but did not associate with AMH or metabolic parameters.
Collapse
Affiliation(s)
- Alexandra E. Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- *Correspondence: Alexandra E. Butler ;
| | - Vimal Ramachandran
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Rhiannon David
- Department of Surgery & Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Nigel J. Gooderham
- Department of Surgery & Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Manasi Benurwar
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Soha R. Dargham
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Shahina Hayat
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - S. Hani Najafi-Shoushtari
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, United States
| | - Stephen L. Atkin
- Division of Research, Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, Doha, Qatar
- Royal College of Surgeons Ireland, Busaiteen, Bahrain
| |
Collapse
|
35
|
Butler AE, Hayat S, Dargham SR, Malek JA, Abdulla SA, Mohamoud YA, Suhre K, Sathyapalan T, Atkin SL. Alterations in long noncoding RNAs in women with and without polycystic ovarian syndrome. Clin Endocrinol (Oxf) 2019; 91:793-797. [PMID: 31482638 DOI: 10.1111/cen.14087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 12/23/2022]
Abstract
UNLABELLED Long noncoding RNAs (lncRNAs) are RNA transcripts over 200 nucleotides long that are not translated into protein; however, there is increasing evidence of their regulatory functions. To date, there are few studies measuring lncRNA in control women or women with polycystic ovary syndrome (PCOS). OBJECTIVE To determine lncRNA differences between PCOS and control women. DESIGN Cross sectional study. PATIENTS Twenty four anovulatory women with all three diagnostic features of PCOS compared to 24 control women in the follicular phase of their menstrual cycle from a PCOS biobank. RESULTS Women with PCOS were age and weight matched compared to the control women but were significantly insulin resistant and hyperandrogenemic (P < .01). Eight lncRNA (P < .05) were detected that differed between PCOS and control women, but only MIRLET7BHG correlated with body mass index (r = .66, P < .05). No lncRNA correlated with antimullerian hormone (AMH) levels, insulin resistance (HOMA-IR) or the free androgen index (FAI). Ingenuity pathway assessment (IPA) did not identify any functional pathways for the lncRNAs. CONCLUSION LncRNAs differ between anovulatory PCOS and control women in the follicular phase of the menstrual cycle. It is unclear if this is due to inherent differences between PCOS and control women or due to changes in lncRNA that are menstrual cycle dependent. However, their IPA did not identify linked pathways, likely because few functions are as yet assigned to these lncRNAs.
Collapse
Affiliation(s)
- Alexandra E Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Shahina Hayat
- Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Soha R Dargham
- Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Joel A Malek
- Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | | | | | - Karsten Suhre
- Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | | | - Stephen L Atkin
- Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Royal College of Surgeons Ireland, Busaiteen, Bahrain
| |
Collapse
|
36
|
Hiam D, Simar D, Laker R, Altıntaş A, Gibson-Helm M, Fletcher E, Moreno-Asso A, Trewin AJ, Barres R, Stepto NK. Epigenetic Reprogramming of Immune Cells in Women With PCOS Impact Genes Controlling Reproductive Function. J Clin Endocrinol Metab 2019; 104:6155-6170. [PMID: 31390009 DOI: 10.1210/jc.2019-01015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is a chronic disease affecting reproductive function and whole-body metabolism. Although the etiology is unclear, emerging evidence indicates that the epigenetics may be a contributing factor. OBJECTIVE To determine the role of global and genome-wide epigenetic modifications in specific immune cells in PCOS compared with controls and whether these could be related to clinical features of PCOS. DESIGN Cross-sectional study. PARTICIPANTS Women with (n = 17) or without PCOS (n = 17). SETTING Recruited from the general community. MAIN OUTCOME MEASURES Isolated peripheral blood mononuclear cells were analyzed using multicolor flow cytometry methods to determine global DNA methylation levels in a cell-specific fashion. Transcriptomic and genome-wide DNA methylation analyses were performed on T helper cells using RNA sequencing and reduced representation bisulfite sequencing. RESULTS Women with PCOS had lower global DNA methylation in monocytes (P = 0.006) and in T helper (P = 0.004), T cytotoxic (P = 0.004), and B cells (P = 0.03). Specific genome-wide DNA methylation analysis of T helper cells from women with PCOS identified 5581 differentially methylated CpG sites. Functional gene ontology enrichment analysis showed that genes located at the proximity of differentially methylated CpG sites belong to pathways related to reproductive function and immune cell function. However, these genes were not altered at the transcriptomic level. CONCLUSIONS It was shown that PCOS is associated with global and gene-specific DNA methylation remodeling in a cell type-specific manner. Further investigation is warranted to determine whether epigenetic reprogramming of immune cells is important in determining the different phenotypes of PCOS.
Collapse
Affiliation(s)
- Danielle Hiam
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Rhianna Laker
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Ali Altıntaş
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Melanie Gibson-Helm
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elly Fletcher
- Baker Heart and Disease Institute, Melbourne, Victoria, Australia
| | - Alba Moreno-Asso
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Adam J Trewin
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Romain Barres
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Nigel K Stepto
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Medicine-Western Health, School of Medicine, Faculty of Medicine, Dentistry, and Health Science, Melbourne, Australia
| |
Collapse
|
37
|
Abbott DH, Kraynak M, Dumesic DA, Levine JE. In utero Androgen Excess: A Developmental Commonality Preceding Polycystic Ovary Syndrome? FRONTIERS OF HORMONE RESEARCH 2019; 53:1-17. [PMID: 31499494 DOI: 10.1159/000494899] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In utero androgen excess reliably induces polycystic ovary syndrome (PCOS)-like reproductive and metabolic traits in female monkeys, sheep, rats, and mice. In humans, however, substantial technical and ethical constraints on fetal sampling have curtailed safe, pathogenic exploration during gestation. Evidence consistent with in utero origins for PCOS in humans has thus been slow to amass, but the balance now leans toward developmental fetal origins. Given that PCOS is familial and highly heritable, difficulties encountered in discerning genetic contributions to PCOS pathogenesis are puzzling and, to date, accounts for <10% of PCOS presentations. Unaccounted heritability notwithstanding, molecular commonality in pathogenic mechanisms is emerging, suggested by co-occurrence at the same gene loci of (1) PCOS genetic variants (PCOS women), (2) epigenetic alterations in DNA methylation (PCOS women), and (3) bioinformatics, gene networks-identified, epigenetic alterations in DNA methylation (female rhesus monkeys exposed to testosterone (T) in utero). In addition, naturally occurring hyperandrogenism in female monkeys singles out individuals with PCOS-like reproductive and metabolic traits accompanied by somatic biomarkers of in utero T exposure. Such phenotypic and molecular convergence between highly related species suggests not only dual genetic and epigenetic contributions to a developmental origin of PCOS but also common molecular pathogenesis extending beyond humans.
Collapse
Affiliation(s)
- David H Abbott
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA, .,Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA, .,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA,
| | - Marissa Kraynak
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA.,Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
38
|
Vázquez-Martínez ER, Gómez-Viais YI, García-Gómez E, Reyes-Mayoral C, Reyes-Muñoz E, Camacho-Arroyo I, Cerbón M. DNA methylation in the pathogenesis of polycystic ovary syndrome. Reproduction 2019; 158:R27-R40. [DOI: 10.1530/rep-18-0449] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the leading endocrine and metabolic disorder in premenopausal women characterized by hyperandrogenism and abnormal development of ovarian follicles. To date, the PCOS etiology remains unclear and has been related to insulin resistance, obesity, type 2 diabetes mellitus, cardiovascular disease and infertility, among other morbidities. Substantial evidence illustrates the impact of genetic, intrauterine and environmental factors on the PCOS etiology. Lately, epigenetic factors have garnered considerable attention in the pathogenesis of PCOS considering that changes in the content of DNA methylation, histone acetylation and noncoding RNAs have been reported in various tissues of women with this disease. DNA methylation is changed in the peripheral and umbilical cord blood, as well as in ovarian and adipose tissue of women with PCOS, suggesting the involvement of this epigenetic modification in the pathogenesis of the disease. Perhaps, these defects in DNA methylation promote the deregulation of genes involved in inflammation, hormone synthesis and signaling and glucose and lipid metabolism. Research on the role of DNA methylation in the pathogenesis of PCOS is just beginning, and several issues await investigation. This review aims to provide an overview of current research focused on DNA methylation and PCOS, as well as discuss the perspectives regarding this topic.
Collapse
|
39
|
Hosseini E, Shahhoseini M, Afsharian P, Karimian L, Ashrafi M, Mehraein F, Afatoonian R. Role of epigenetic modifications in the aberrant CYP19A1 gene expression in polycystic ovary syndrome. Arch Med Sci 2019; 15:887-895. [PMID: 31360184 PMCID: PMC6657255 DOI: 10.5114/aoms.2019.86060] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/23/2017] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION In this study, the global DNA methylation, histone acetylation and methylation levels of cumulus cells (CCs) in infertile polycystic ovary syndrome (PCOS) patients and the correlation of these epigenetic modifications with the expression of the ovarian aromatase gene (as an important marker in the etiology of PCOS) were investigated. MATERIAL AND METHODS A cross-sectional study was conducted on 24 patients (12 PCOS patients and 12 healthy women), who underwent ovarian stimulation. Nucleosome ELISA was performed, in order to identify the global occupancy level of Mecp2 (as a marker of DNA methylation) and H3K9me2/H3K9ac as histone modification markers in chromatin fractions obtained from CCs. The CYP19A1 gene expression was measured by qRT-PCR. The level of DNA incorporation of MeCP2, histone modification markers and binding of estrogen receptor β (ERβ) to CYP19A1 regulatory sequences were examined by ChIP-QPCR assay. RESULTS The data demonstrate a significant increase in global occupancy levels of MeCP2 and H3K9ac markers and a decrease of H3K9me2 to chromatin in CCs of PCOS patients vs. control group. Furthermore, CYP19A1 gene expression, and the incorporation of H3K9ac in PII, PI.3, and PI.4 promoters of CYP19A1 in PCOS, were higher than those of controls. Also, significant hypomethylation of H3K9 at PII and DNA hypomethylated at PII and PI.3 promoters and differential binding of ERβ to three promoters were observed in PCOS patients (p < 0.05). CONCLUSIONS Aromatase expression can be affected by epigenetic modifications and differential ERβ binding to the proximal CYP19A1 promoters. These mechanisms may be involved in the enhanced aromatase transcription during ovarian stimulation in PCOS patients.
Collapse
Affiliation(s)
- Elham Hosseini
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Obstetrics and Gynecology, IVF Clinic, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Shahhoseini
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Parvaneh Afsharian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Leila Karimian
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahnaz Ashrafi
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Afatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
40
|
Ribas L, Crespo B, Sánchez-Baizán N, Xavier D, Kuhl H, Rodríguez JM, Díaz N, Boltañá S, MacKenzie S, Morán F, Zanuy S, Gómez A, Piferrer F. Characterization of the European Sea Bass (Dicentrarchus labrax) Gonadal Transcriptome During Sexual Development. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:359-373. [PMID: 30919121 DOI: 10.1007/s10126-019-09886-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
The European sea bass is one of the most important cultured fish in Europe and has a marked sexual growth dimorphism in favor of females. It is a gonochoristic species with polygenic sex determination, where a combination between still undifferentiated genetic factors and environmental temperature determines sex ratios. The molecular mechanisms responsible for gonadal sex differentiation are still unknown. Here, we sampled fish during the gonadal developmental period (110 to 350 days post fertilization, dpf), and performed a comprehensive transcriptomic study by using a species-specific microarray. This analysis uncovered sex-specific gonadal transcriptomic profiles at each stage of development, identifying larger number of differentially expressed genes in ovaries when compared to testis. The expression patterns of 54 reproduction-related genes were analyzed. We found that hsd17β10 is a reliable marker of early ovarian differentiation. Further, three genes, pdgfb, snx1, and nfy, not previously related to fish sex differentiation, were tightly associated with testis development in the sea bass. Regarding signaling pathways, lysine degradation, bladder cancer, and NOD-like receptor signaling were enriched for ovarian development while eight pathways including basal transcription factors and steroid biosynthesis were enriched for testis development. Analysis of the transcription factor abundance showed an earlier increase in females than in males. Our results show that, although many players in the sex differentiation pathways are conserved among species, there are peculiarities in gene expression worth exploring. The genes identified in this study illustrate the diversity of players involved in fish sex differentiation and can become potential biomarkers for the management of sex ratios in the European sea bass and perhaps other cultured species.
Collapse
Affiliation(s)
- L Ribas
- Institute of Marine Sciences (ICM), Spanish National Research Council (CSIC), Barcelona, Catalonia, Spain
| | - B Crespo
- Institute of Aquaculture of Torre de la Sal (IATS-CSIC), Ribera de Cabanes s/n. Torre la Sal, 12595, Castellón, Spain
- UCL GOS Institute of Child Health, University College London, London, UK
| | - N Sánchez-Baizán
- Institute of Marine Sciences (ICM), Spanish National Research Council (CSIC), Barcelona, Catalonia, Spain
| | - D Xavier
- Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain
| | - H Kuhl
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Ecophysiology and Aquaculture, Leibniz Institute for Freshwater Ecology and Inland Fisheries, Berlin, Germany
| | - J M Rodríguez
- Spanish National Bioinformatics Institute, Madrid, Spain
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - N Díaz
- Institute of Marine Sciences (ICM), Spanish National Research Council (CSIC), Barcelona, Catalonia, Spain
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - S Boltañá
- Autonomous University of Barcelona, Barcelona, Spain
- Interdisciplinary Center for Aquaculture Research (INCAR), Department of Oceanography, Biotechnology Center, University of Concepción, Concepción, Chile
| | - S MacKenzie
- Autonomous University of Barcelona, Barcelona, Spain
- Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK
| | - F Morán
- Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain
| | - S Zanuy
- Institute of Aquaculture of Torre de la Sal (IATS-CSIC), Ribera de Cabanes s/n. Torre la Sal, 12595, Castellón, Spain
| | - A Gómez
- Institute of Aquaculture of Torre de la Sal (IATS-CSIC), Ribera de Cabanes s/n. Torre la Sal, 12595, Castellón, Spain.
| | - F Piferrer
- Institute of Marine Sciences (ICM), Spanish National Research Council (CSIC), Barcelona, Catalonia, Spain.
| |
Collapse
|
41
|
Sagvekar P, Kumar P, Mangoli V, Desai S, Mukherjee S. DNA methylome profiling of granulosa cells reveals altered methylation in genes regulating vital ovarian functions in polycystic ovary syndrome. Clin Epigenetics 2019; 11:61. [PMID: 30975191 PMCID: PMC6458760 DOI: 10.1186/s13148-019-0657-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Background Women with polycystic ovary syndrome (PCOS) manifest a host of ovarian defects like impaired folliculogenesis, anovulation, and poor oocyte quality, which grossly affect their reproductive health. Addressing the putative epigenetic anomalies that tightly regulate these events is of foremost importance in this disorder. We therefore aimed to carry out DNA methylome profiling of cumulus granulosa cells and assess the methylation and transcript expression profiles of a few differentially methylated genes contributing to ovarian defects in PCOS. A total of 20 controls and 20 women with PCOS were selected from a larger cohort of women undergoing IVF, after carefully screening their sera and follicular fluids for hormonal and biochemical parameters. DNA extracted from cumulus granulosa cells of three women each, from control and PCOS groups was subjected to high-throughput, next generation bisulfite sequencing, using the Illumina HiSeq 2500® platform. Remaining samples were used for the validation of methylation status of some identified genes by pyrosequencing, and the transcript expression profiles of these genes were assessed by quantitative real-time PCR. Results In all, 6486 CpG sites representing 3840 genes associated with Wnt signaling, G protein receptor, endothelin/integrin signaling, angiogenesis, chemokine/cytokine-mediated inflammation, etc., showed differential methylation in PCOS. Hypomethylation was noted in 2977 CpGs representing 2063 genes while 2509 CpGs within 1777 genes showed hypermethylation. Methylation differences were also noted in noncoding RNAs regulating several ovarian functions that are dysregulated in PCOS. Few differentially methylated genes such as aldo-keto reductase family 1 member C3, calcium-sensing receptor, resistin, mastermind-like domain 1, growth hormone-releasing hormone receptor and tumor necrosis factor, which predominantly contribute to hyperandrogenism, premature luteolysis, and oocyte development defects, were explored as novel epigenetic candidates in mediating ovarian dysfunction. Methylation profiles of these genes matched with our NGS findings, and their transcript expression patterns correlated with the gene hypo- or hypermethylation status. Conclusion Our findings suggest that the epigenetic dysregulation of genes involved in important processes associated with follicular development may contribute to ovarian defects observed in women with PCOS. Electronic supplementary material The online version of this article (10.1186/s13148-019-0657-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pooja Sagvekar
- Department of Molecular Endocrinology, ICMR-National Institute for Research in Reproductive Health, J.M. Street, Parel, Mumbai, Maharashtra, 400012, India
| | - Pankaj Kumar
- Colin Jamura Lab, Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), GKVK, Bellary Road, Bangalore, 560065, India
| | - Vijay Mangoli
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai, Maharashtra, 400007, India
| | - Sadhana Desai
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai, Maharashtra, 400007, India
| | - Srabani Mukherjee
- Department of Molecular Endocrinology, ICMR-National Institute for Research in Reproductive Health, J.M. Street, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
42
|
Abbott DH, Dumesic DA, Levine JE. Hyperandrogenic origins of polycystic ovary syndrome - implications for pathophysiology and therapy. Expert Rev Endocrinol Metab 2019; 14:131-143. [PMID: 30767580 PMCID: PMC6992448 DOI: 10.1080/17446651.2019.1576522] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) diagnosis comprises combinations of female hyperandrogenism, menstrual irregularity and polycystic ovaries. While it is a familial and highly prevalent endocrine disorder, progress towards a cure is hindered by absence of a definitive pathogenic mechanism and lack of an animal model of naturally occurring PCOS. AREAS COVERED These include an overview of PCOS and its potential etiology, and an examination of insights gained into its pathogenic origins. Animal models derived from experimentally-induced hyperandrogenism during gestation, or from naturally-occurring PCOS-like traits, most reliably demonstrate reproductive, neuroendocrine and metabolic pathogenesis. EXPERT OPINION Genetic studies, while identifying at least 17 PCOS risk genes, account for <10% of women with PCOS. A number of PCOS risk genes involve regulation of gonadotropin secretion or action, suggesting a reproductive neuroendocrine basis for PCOS pathogenesis. Consistent with this notion, a number of animal models employing fetal androgen excess demonstrate epigenetic induction of PCOS-like traits, including reproductive neuroendocrine and metabolic dysfunction. Monkey models are most comprehensive, while mouse models provide molecular insight, including identifying the androgen receptor, particularly in neurons, as mediating androgen-induced PCOS-like programming. Naturally-occurring female hyperandrogenism is also demonstrated in monkeys. Animal models are poised to delineate molecular gateways to PCOS pathogenesis.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jon E Levine
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
43
|
Silencing of LncRNA steroid receptor RNA activator attenuates polycystic ovary syndrome in mice. Biochimie 2019; 157:48-56. [DOI: 10.1016/j.biochi.2018.10.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/12/2018] [Indexed: 01/21/2023]
|
44
|
Cui P, Ma T, Tamadon A, Han S, Li B, Chen Z, An X, Shao LR, Wang Y, Feng Y. Hypothalamic DNA methylation in rats with dihydrotestosterone-induced polycystic ovary syndrome: effects of low-frequency electro-acupuncture. Exp Physiol 2018; 103:1618-1632. [PMID: 30204276 DOI: 10.1113/ep087163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/10/2018] [Indexed: 04/30/2025]
Abstract
NEW FINDINGS What is the central question of this study? What is the role of hypothalamic DNA methylation in the development of polycystic ovary syndrome (PCOS) and the response to electro-acupuncture treatment. What is the main finding and its importance? Global DNA methylation and expression of DNA methyltransferases (DNMTs) were increased in PCOS-like rats, and electro-acupuncture (EA) decreased global DNA methylation and DNMT3b expression. Pyrosequencing showed that the DNA methylation of some PCOS candidate genes was changed in the PCOS and PCOS+EA groups, suggesting that hypothalamic DNA methylation plays an important role in the development of PCOS and in mediating the effects of electro-acupuncture treatment. ABSTRACT Polycystic ovary syndrome (PCOS) is a common reproductive and endocrine disease of unknown aetiology. Recently, epigenetic studies focusing on DNA methylation in PCOS have received much attention, but the mechanisms are still unclear. In the present study, we used the 5α-dihydrotestosterone-induced PCOS-like rat model and treated the rats with electro-acupuncture (EA). Rats were randomly divided into four groups - controls, diet-induced obesity, PCOS and PCOS+EA. We examined the reproductive, metabolic and behavioural phenotypes, validated the effect of EA, and explored the role of hypothalamic DNA methylation by analysing the methylation of global DNA and selected candidate genes. The PCOS rats presented with reproductive dysfunctions such as lack of regular oestrous cyclicity, metabolic disorders such as increased body weight and insulin resistance, and depression and anxiety-like behaviours. EA improved the reproductive functions, decreased body weight and improved experimental depressive behaviour. Furthermore, global DNA methylation and the expression of DNA methyltransferases (DNMTs) were increased in PCOS rats compared to the control group, and EA decreased the global DNA methylation and the expression of DNMT3b. In addition, pyrosequencing showed that the DNA methylation of certain CpG sites in targeted genes (Plcg1, Camk2b, Esr2 and Pgr) was increased in the PCOS group, but the DNA methylation of Camk2b and Ar was decreased after EA treatment. These results indicate that hypothalamic DNA methylation might be correlated with the development of PCOS and that EA has an effect on hypothalamic DNA methylation in PCOS rats.
Collapse
Affiliation(s)
- Peng Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Tong Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Amin Tamadon
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Sha Han
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Bing Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Zheyi Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Xiaofei An
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function
| |
Collapse
|
45
|
Ibáñez L, Oberfield SE, Witchel S, Auchus RJ, Chang RJ, Codner E, Dabadghao P, Darendeliler F, Elbarbary NS, Gambineri A, Garcia Rudaz C, Hoeger KM, López-Bermejo A, Ong K, Peña AS, Reinehr T, Santoro N, Tena-Sempere M, Tao R, Yildiz BO, Alkhayyat H, Deeb A, Joel D, Horikawa R, de Zegher F, Lee PA. An International Consortium Update: Pathophysiology, Diagnosis, and Treatment of Polycystic Ovarian Syndrome in Adolescence. Horm Res Paediatr 2018; 88:371-395. [PMID: 29156452 DOI: 10.1159/000479371] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022] Open
Abstract
This paper represents an international collaboration of paediatric endocrine and other societies (listed in the Appendix) under the International Consortium of Paediatric Endocrinology (ICPE) aiming to improve worldwide care of adolescent girls with polycystic ovary syndrome (PCOS)1. The manuscript examines pathophysiology and guidelines for the diagnosis and management of PCOS during adolescence. The complex pathophysiology of PCOS involves the interaction of genetic and epigenetic changes, primary ovarian abnormalities, neuroendocrine alterations, and endocrine and metabolic modifiers such as anti-Müllerian hormone, hyperinsulinemia, insulin resistance, adiposity, and adiponectin levels. Appropriate diagnosis of adolescent PCOS should include adequate and careful evaluation of symptoms, such as hirsutism, severe acne, and menstrual irregularities 2 years beyond menarche, and elevated androgen levels. Polycystic ovarian morphology on ultrasound without hyperandrogenism or menstrual irregularities should not be used to diagnose adolescent PCOS. Hyperinsulinemia, insulin resistance, and obesity may be present in adolescents with PCOS, but are not considered to be diagnostic criteria. Treatment of adolescent PCOS should include lifestyle intervention, local therapies, and medications. Insulin sensitizers like metformin and oral contraceptive pills provide short-term benefits on PCOS symptoms. There are limited data on anti-androgens and combined therapies showing additive/synergistic actions for adolescents. Reproductive aspects and transition should be taken into account when managing adolescents.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Endocrinology, Hospital Sant Joan de Deu, Esplugues, Barcelona, Spain.,CIBERDEM, ISCIII, Madrid, Spain
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Selma Witchel
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | | | - R Jeffrey Chang
- Department of Reproductive Medicine, UCSD School of Medicine, La Jolla, California, USA
| | - Ethel Codner
- Institute of Maternal and Child Research, University of Chile, School of Medicine, Santiago, Chile
| | - Preeti Dabadghao
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | - Alessandra Gambineri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cecilia Garcia Rudaz
- Division of Women, Youth and Children, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kathleen M Hoeger
- Department of OBGYN, University of Rochester Medical Center, Rochester, New York, USA
| | - Abel López-Bermejo
- Pediatric Endocrinology, Hospital de Girona Dr. Josep Trueta, Girona, Spain
| | - Ken Ong
- MRC Epidemiology Unit, University of Cambridge, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Alexia S Peña
- The University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia
| | - Thomas Reinehr
- University of Witten/Herdecke, Vestische Kinder- und Jugendklinik, Pediatric Endocrinology, Diabetes, and Nutrition Medicine, Datteln, Germany
| | - Nicola Santoro
- Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Rachel Tao
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Bulent O Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| | - Haya Alkhayyat
- Medical University of Bahrain, BDF Hospital, Riffa, Bahrein
| | - Asma Deeb
- Mafraq Hospital, Abu Dhabi, United Arab Emirates
| | - Dipesalema Joel
- Department of Paediatrics and Adolescent Health, University of Botswana Teaching Hospital, Gaborone, Botswana
| | - Reiko Horikawa
- Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Francis de Zegher
- Department Pediatrics, University Hospital Gasthuisberg, Leuven, Belgium
| | - Peter A Lee
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
46
|
Goldrat O, Delbaere A. PCOS: update and diagnostic approach. Clin Biochem 2018; 62:24-31. [PMID: 30195483 DOI: 10.1016/j.clinbiochem.2018.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/26/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Oranite Goldrat
- Fertility Clinic, Department of Obstetrics and Gynecology, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Route de Lennik, 808, Brussels, Belgium.
| | - Anne Delbaere
- Fertility Clinic, Department of Obstetrics and Gynecology, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Route de Lennik, 808, Brussels, Belgium.
| |
Collapse
|
47
|
Montazerian M, Yasari F, Aghaalikhani N. Ovarian extracellular MicroRNAs as the potential non-invasive biomarkers: An update. Biomed Pharmacother 2018; 106:1633-1640. [PMID: 30119239 DOI: 10.1016/j.biopha.2018.07.073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 01/06/2023] Open
Abstract
Through the reproductive system, it has been realized that the microRNAs (miRNAs) have emerged as one of the principal post-transcriptional gene regulators of the diverse developmental processes. The ovary, as a dynamic organ, co-ordinates follicle recruitment, selection, and ovulation, in which miRNAs play the central role almost in its all functions. Deregulation of these developmental procedures in ovary could lead to the ovarian dysfunction, infertility, decrease in the assisted reproductive treatment (ART) outcome, and death in some patients with ovarian cancer. In recent years, detection of ovarian extracellular miRNAs in body fluids such as follicular fluid and serum/plasma has opened a new era in the biomarker discovery field. Here through the present review, different aspects of the potential and proposed involvement of the extracellular miRNAs in both physiologic and pathologic contexts of the ovary have been discussed. Moreover, the researchers have addressed the relevant findings, challenges, and issues which associated with the extracellular miRNAs in the ovarian microenvironments to provide the better insight into understanding the molecular mechanisms which were involved in the pathophysiologic conditions. Finally, a comprehensive survey of the gaps has been discussed to hopefully shed new light and perspective on the development of the novel diagnostic and therapeutic platforms in the clinic.
Collapse
Affiliation(s)
- Mojgan Montazerian
- Department of Midwifery, Dezful Branch Islamic Azad University, Dezful, Iran.
| | - Fahimeh Yasari
- Department of Midwifery, Dezful Branch Islamic Azad University, Dezful, Iran
| | - Nazi Aghaalikhani
- Department of Midwifery, Dezful Branch Islamic Azad University, Dezful, Iran
| |
Collapse
|
48
|
Su NJ, Ma J, Feng DF, Zhou S, Li ZT, Zhou WP, Deng H, Liang JY, Yang XH, Zhang YM, Liu FH, Zhang L. The peripheral blood transcriptome identifies dysregulation of inflammatory response genes in polycystic ovary syndrome. Gynecol Endocrinol 2018; 34:584-588. [PMID: 29262729 DOI: 10.1080/09513590.2017.1418851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder in women, resulting in ovulation failure and other metabolic problems. However, the underlying mechanisms of it remain largely uncertain due to the complexity of clinical manifestations. This systemic disorder is involved in endocrine, metabolism, immune system and many organs, and few studies have explored peripheral blood transcriptome in patients with PCOS. We performed gene expression profiling of peripheral blood from 8 PCOS patients and eight healthy women with microarray. The significance analysis of microarray (SAM) software was employed to screen the differentially expressed genes (DEGs) and gene ontology (GO) was used for functional enrichment analysis. In total, 181 DEGs with fold-changes >2.0 and q-values <0.05 were identified between the two groups. Among them, 149 were up-regulated and 32 down-regulated in PCOS. Unsupervised clustering of expressed genes could readily differentiate PCOS from control. More importantly, inflammatory response pathway including 14 dysregulated genes was highly enriched in PCOS. Furthermore, 10 DEGs were validated using quantitative reverse-transcription PCR (qRT-PCR) assays. Our study provides independent evidence for the involvement of systemic inflammatory response in PCOS and it may facilitate a greater understanding of this complex disease.
Collapse
Affiliation(s)
- Nian-Jun Su
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Jian Ma
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - De-Feng Feng
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Shuai Zhou
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Zi-Tao Li
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Wei-Ping Zhou
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Hua Deng
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Jia-Ying Liang
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Xu-Hui Yang
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Yue-Mei Zhang
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Feng-Hua Liu
- a Reproductive Medical Center , Guangdong Women and Children Hospital , Guangzhou , China
| | - Liang Zhang
- b Translational Medicine Center , Guangdong Women and Children Hospital , Guangzhou , China
| |
Collapse
|
49
|
Pan JX, Tan YJ, Wang FF, Hou NN, Xiang YQ, Zhang JY, Liu Y, Qu F, Meng Q, Xu J, Sheng JZ, Huang HF. Aberrant expression and DNA methylation of lipid metabolism genes in PCOS: a new insight into its pathogenesis. Clin Epigenetics 2018; 10:6. [PMID: 29344314 PMCID: PMC5767000 DOI: 10.1186/s13148-018-0442-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/08/2018] [Indexed: 11/10/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS), whose etiology remains uncertain, is a highly heterogenous and genetically complex endocrine disorder. The aim of this study was to identify differentially expressed genes (DEGs) in granulosa cells (GCs) from PCOS patients and make epigenetic insights into the pathogenesis of PCOS. Results Included in this study were 110 women with PCOS and 119 women with normal ovulatory cycles undergoing in vitro fertilization acting as the control group. RNA-seq identified 92 DEGs unique to PCOS GCs in comparison with the control group. Bioinformatic analysis indicated that synthesis of lipids and steroids was activated in PCOS GCs. 5-Methylcytosine analysis demonstrated that there was an approximate 25% reduction in global DNA methylation of GCs in PCOS women (4.44 ± 0.65%) compared with the controls (6.07 ± 0.72%; P < 0.05). Using MassArray EpiTYPER quantitative DNA methylation analysis, we also found hypomethylation of several gene promoters related to lipid and steroid synthesis, which might result in the aberrant expression of these genes. Conclusions Our results suggest that hypomethylated genes related to the synthesis of lipid and steroid may dysregulate expression of these genes and promote synthesis of steroid hormones including androgen, which could partially explain mechanisms of hyperandrogenism in PCOS. Electronic supplementary material The online version of this article (10.1186/s13148-018-0442-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie-Xue Pan
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China.,2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China.,4Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
| | - Ya-Jing Tan
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Fang-Fang Wang
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| | - Ning-Ning Hou
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| | - Yu-Qian Xiang
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Jun-Yu Zhang
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Ye Liu
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Fan Qu
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| | - Qing Meng
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| | - Jian Xu
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| | - Jian-Zhong Sheng
- 2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China.,3Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, 310058 China
| | - He-Feng Huang
- 1The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China.,2The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, Zhejiang 310006 China
| |
Collapse
|
50
|
Xu J, Bao X, Peng Z, Wang L, Du L, Niu W, Sun Y. Comprehensive analysis of genome-wide DNA methylation across human polycystic ovary syndrome ovary granulosa cell. Oncotarget 2017; 7:27899-909. [PMID: 27056885 PMCID: PMC5053696 DOI: 10.18632/oncotarget.8544] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/28/2016] [Indexed: 01/11/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) affects approximately 7% of the reproductive-age women. A growing body of evidence indicated that epigenetic mechanisms contributed to the development of PCOS. The role of DNA modification in human PCOS ovary granulosa cell is still unknown in PCOS progression. Global DNA methylation and hydroxymethylation were detected between PCOS’ and controls’ granulosa cell. Genome-wide DNA methylation was profiled to investigate the putative function of DNA methylaiton. Selected genes expressions were analyzed between PCOS’ and controls’ granulosa cell. Our results showed that the granulosa cell global DNA methylation of PCOS patients was significant higher than the controls’. The global DNA hydroxymethylation showed low level and no statistical difference between PCOS and control. 6936 differentially methylated CpG sites were identified between control and PCOS-obesity. 12245 differential methylated CpG sites were detected between control and PCOS-nonobesity group. 5202 methylated CpG sites were significantly differential between PCOS-obesity and PCOS-nonobesity group. Our results showed that DNA methylation not hydroxymethylation altered genome-wide in PCOS granulosa cell. The different methylation genes were enriched in development protein, transcription factor activity, alternative splicing, sequence-specific DNA binding and embryonic morphogenesis. YWHAQ, NCF2, DHRS9 and SCNA were up-regulation in PCOS-obesity patients with no significance different between control and PCOS-nonobesity patients, which may be activated by lower DNA methylaiton. Global and genome-wide DNA methylation alteration may contribute to different genes expression and PCOS clinical pathology.
Collapse
Affiliation(s)
- Jiawei Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiao Bao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhaofeng Peng
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Linlin Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Linqing Du
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Wenbin Niu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| |
Collapse
|