1
|
Li J, Fan L, Wei J, Huang W. Elucidating the pathophysiology of polycystic ovary syndrome: Construction and analysis of a ceRNA network in cumulus cells. Reprod Biol 2025; 25:100916. [PMID: 39566253 DOI: 10.1016/j.repbio.2024.100916] [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: 03/13/2024] [Revised: 05/13/2024] [Accepted: 06/16/2024] [Indexed: 11/22/2024]
Abstract
Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder with elusive molecular mechanisms. This study explores the competitive endogenous RNA (ceRNA) regulatory network in the cumulus cells of PCOS patients. ceRNAs are transcripts like mRNAs, miRNAs, and lncRNAs that competitively bind shared miRNAs, regulating gene expression post-transcriptionally. We analyzed mRNA, microRNA (miRNA), and long non-coding RNA (lncRNA) from two cohorts: 12 PCOS patients and 11 healthy controls (dataset GSE10946), and 5 PCOS patients and 5 healthy controls (dataset GSE72274). These microarray datasets, obtained from the Gene Expression Omnibus (GEO), helped us identify differentially expressed mRNAs, miRNAs, and lncRNAs. Our analysis revealed a significant ceRNA network, which may play a crucial role in the pathophysiology of PCOS. In this network, 5 lncRNAs, 3 miRNAs, and 36 mRNAs were identified as differentially expressed. These elements form a complex regulatory schema influencing key cellular processes related to the disease, such as cell cycle regulation and response to estrogen. The HOXA11-AS-hsa-miR-454-3p-CCND2 network emerged as a potentially valuable biomarker for PCOS diagnosis, supported by Receiver Operating Characteristic (ROC) curve analysis indicating strong predictive power. Our findings suggest that the ceRNA interactions in PCOS cumulus cells provide a deeper understanding of the disease's molecular basis and offer new avenues for therapeutic intervention. This in silico study lays the groundwork for further experimental validation of these ceRNA networks as targets for PCOS treatment.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Li Fan
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Jiajia Wei
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Wenjie Huang
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China.
| |
Collapse
|
2
|
Jo M, Brännström M, Akins JW, Curry TE. New insights into the ovulatory process in the human ovary. Hum Reprod Update 2025; 31:21-47. [PMID: 39331957 PMCID: PMC11696709 DOI: 10.1093/humupd/dmae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Successful ovulation is essential for natural conception and fertility. Defects in the ovulatory process are associated with various conditions of infertility or subfertility in women. However, our understanding of the intra-ovarian biochemical mechanisms underlying this process in women has lagged compared to our understanding of animal models. This has been largely due to the limited availability of human ovarian samples that can be used to examine changes across the ovulatory period and delineate the underlying cellular/molecular mechanisms in women. Despite this challenge, steady progress has been made to improve our knowledge of the ovulatory process in women by: (i) collecting granulosa cells across the IVF interval, (ii) creating a novel approach to collecting follicular cells and tissues across the periovulatory period from normally cycling women, and (iii) developing unique in vitro models to examine the LH surge or hCG administration-induced ovulatory changes in gene expression, the regulatory mechanisms underlying the ovulatory changes, and the specific functions of the ovulatory factors. OBJECTIVE AND RATIONALE The objective of this review is to summarize findings generated using in vivo and in vitro models of human ovulation, with the goal of providing new insights into the mechanisms underlying the ovulatory process in women. SEARCH METHODS This review is based on the authors' own studies and a search of the relevant literature on human ovulation to date using PubMed search terms such as 'human ovulation EGF-signaling', 'human ovulation steroidogenesis', 'human ovulation transcription factor', 'human ovulation prostaglandin', 'human ovulation proteinase', 'human ovulation angiogenesis' 'human ovulation chemokine', 'human ovulatory disorder', 'human granulosa cell culture'. Our approach includes comparing the data from the authors' studies with the existing microarray or RNA-seq datasets generated using ovarian cells obtained throughout the ovulatory period from humans, monkeys, and mice. OUTCOMES Current findings from studies using in vivo and in vitro models demonstrate that the LH surge or hCG administration increases the expression of ovulatory mediators, including EGF-like factors, steroids, transcription factors, prostaglandins, proteolytic systems, and other autocrine and paracrine factors, similar to those observed in other animal models such as rodents, ruminants, and monkeys. However, the specific ovulatory factors induced, their expression pattern, and their regulatory mechanisms vary among different species. These species-specific differences stress the necessity of utilizing human samples to delineate the mechanisms underlying the ovulatory process in women. WIDER IMPLICATIONS The data from human ovulation in vivo and in vitro models have begun to fill the gaps in our understanding of the ovulatory process in women. Further efforts are needed to discover novel ovulatory factors. One approach to address these gaps is to improve existing in vitro models to more closely mimic in vivo ovulatory conditions in humans. This is critically important as the knowledge obtained from these human studies can be translated directly to aid in the diagnosis of ovulation-associated pathological conditions, for the development of more effective treatment to help women with anovulatory infertility or, conversely, to better manage ovulation for contraceptive purposes. REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | | | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
3
|
Omidvar-Mehrabadi A, Ebrahimi F, Shahbazi M, Mohammadnia-Afrouzi M. Cytokine and chemokine profiles in women with endometriosis, polycystic ovary syndrome, and unexplained infertility. Cytokine 2024; 178:156588. [PMID: 38555853 DOI: 10.1016/j.cyto.2024.156588] [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: 01/10/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Numerous factors (including immunological, congenital, hormonal, and morphological disorders) can lead to infertility. In this regard, 3 specific diseases associated with infertility are discussed in this review study (i.e., polycystic ovary syndrome [PCOS], endometriosis [EMS], and unexplained infertility [UI]). PCOS is a common endocrine disorder characterized by chronic low-grade inflammation, and EMS is a benign disease characterized by the presence of ectopic endometrial tissue. UI refers to couples who are unable to conceive for no known reason. Conception and pregnancy are significantly affected by the immune system; in this regard, chemokines and cytokines play important roles in the regulation of immune responses. Patients with PCOS, EMS, and UI have altered cytokine and chemokine profiles, suggesting that dysregulation of these molecules may contribute to infertility in these conditions. Accordingly, the issue of infertility is addressed in this review study, a condition that affects approximately 16% of couples worldwide.
Collapse
Affiliation(s)
| | - Fateme Ebrahimi
- Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Shahbazi
- Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | | |
Collapse
|
4
|
Froment P, Plotton I, Giulivi C, Fabre S, Khoueiry R, Mourad NI, Horman S, Ramé C, Rouillon C, Grandhaye J, Bigot Y, Chevaleyre C, Le Guevel R, Mallegol P, Andriantsitohaina R, Guerif F, Tamburini J, Viollet B, Foretz M, Dupont J. At the crossroads of fertility and metabolism: the importance of AMPK-dependent signaling in female infertility associated with hyperandrogenism. Hum Reprod 2022; 37:1207-1228. [PMID: 35459945 DOI: 10.1093/humrep/deac067] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/01/2022] [Indexed: 03/25/2024] Open
Abstract
STUDY QUESTION What biological processes are linked to the signaling of the energy sensor 5'-AMP-activated protein kinase (AMPK) in mouse and human granulosa cells (GCs)? SUMMARY ANSWER The lack of α1AMPK in GCs impacted cell cycle, adhesion, lipid metabolism and induced a hyperandrogenic response. WHAT IS KNOWN ALREADY AMPK is expressed in the ovarian follicle, and its activation by pharmacological medications, such as metformin, inhibits the production of steroids. Polycystic ovary syndrome (PCOS) is responsible for infertility in approximately 5-20% of women of childbearing age and possible treatments include reducing body weight, improving lifestyle and the administration of a combination of drugs to improve insulin resistance, such as metformin. STUDY DESIGN, SIZE, DURATION AMPK signaling was evaluated by analyzing differential gene expression in immortalized human granulosa cells (KGNs) with and without silencing α1AMPK using CRISPR/Cas9. In vivo studies included the use of a α1AMPK knock-out mouse model to evaluate the role of α1AMPK in folliculogenesis and fertility. Expression of α1AMPK was evaluated in primary human granulosa-luteal cells retrieved from women undergoing IVF with and without a lean PCOS phenotype (i.e. BMI: 18-25 kg/m2). PARTICIPANTS/MATERIALS, SETTING, METHODS α1AMPK was disrupted in KGN cells and a transgenic mouse model. Cell viability, proliferation and metabolism were evaluated. Androgen production was evaluated by analyzing protein levels of relevant enzymes in the steroid pathway by western blots, and steroid levels obtained from in vitro and in vivo models by mass spectrometry. Differential gene expression in human GC was obtained by RNA sequencing. Analysis of in vivo murine folliculogenesis was performed by histology and immunochemistry, including evaluation of the anti-Müllerian hormone (AMH) marker. The α1AMPK gene expression was evaluated by quantitative RT-PCR in primary GCs obtained from women with the lean PCOS phenotype (n = 8) and without PCOS (n = 9). MAIN RESULTS AND THE ROLE OF CHANCE Silencing of α1AMPK in KGN increased cell proliferation (P < 0.05 versus control, n = 4), promoted the use of fatty acids over glucose, and induced a hyperandrogenic response resulting from upregulation of two of the enzymes involved in steroid production, namely 3β-hydroxysteroid dehydrogenase (3βHSD) and P450 side-chain cleavage enzyme (P450scc) (P < 0.05, n = 3). Female mice deficient in α1AMPK had a 30% decrease in their ovulation rate (P < 0.05, n = 7) and litter size, a hyperandrogenic response (P < 0.05, n = 7) with higher levels of 3βHSD and p450scc levels in the ovaries, and an increase in the population of antral follicles (P < 0.01, n = 10) compared to controls. Primary GCs from lean women with PCOS had lower α1AMPK mRNA expression levels than the control group (P < 0.05, n = 8-9). LARGE SCALE DATA The FastQ files and metadata were submitted to the European Nucleotide Archive (ENA) at EMBL-EBI under accession number PRJEB46048. LIMITATIONS, REASONS FOR CAUTION The human KGN is a not fully differentiated, transformed cell line. As such, to confirm the role of AMPK in GC and the PCOS phenotype, this model was compared to two others: an α1AMPK transgenic mouse model and primary differentiated granulosa-lutein cells from non-obese women undergoing IVF (with and without PCOS). A clear limitation is the small number of patients with PCOS utilized in this study and that the collection of human GCs was performed after hormonal stimulation. WIDER IMPLICATIONS OF THE FINDINGS Our results reveal that AMPK is directly involved in steroid production in human GCs. In addition, AMPK signaling was associated with other processes frequently reported as dysfunctional in PCOS models, such as cell adhesion, lipid metabolism and inflammation. Silencing of α1AMPK in KGN promoted folliculogenesis, with increases in AMH. Evaluating the expression of the α1AMPK subunit could be considered as a marker of interest in infertility cases related to hormonal imbalances and metabolic disorders, including PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was financially supported by the Institut National de la Recherche Agronomique (INRA) and the national programme « FERTiNERGY » funded by the French National Research Agency (ANR). The authors report no intellectual or financial conflicts of interest related to this work. R.K. is identified as personnel of the International Agency for Research on Cancer/World Health Organization. R.K. alone is responsible for the views expressed in this article and she does not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/World Health Organization. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Pascal Froment
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Ingrid Plotton
- Molecular Endocrinology and Rare Diseases, University Hospital, Claude Bernard Lyon 1 University, Bron, France
| | - Cecilia Giulivi
- Department of Molecular Biosciences, University of California Davis, School of Veterinary Medicine, Davis, CA, USA
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, USA
| | - Stephane Fabre
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Rita Khoueiry
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Nizar I Mourad
- Pôle de Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Christelle Ramé
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | | | - Yves Bigot
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Remy Le Guevel
- Plate-forme ImPACcell, Université de Rennes 1, Rennes, France
| | - Patricia Mallegol
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | | | - Jérôme Tamburini
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Joelle Dupont
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| |
Collapse
|
5
|
Karakaya C, Çil AP, Bilguvar K, Çakir T, Karalok MH, Karabacak RO, Caglayan AO. Further delineation of familial polycystic ovary syndrome (PCOS) via whole-exome sequencing: PCOS-related rare FBN3 and FN1 gene variants are identified. J Obstet Gynaecol Res 2022; 48:1202-1211. [PMID: 35141985 PMCID: PMC9050819 DOI: 10.1111/jog.15187] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/19/2022] [Accepted: 01/29/2022] [Indexed: 12/11/2022]
Abstract
AIM To identify pathogenic rare coding Mendelian/high-effect size variant(s) by whole-exome sequencing in familial polycystic ovary syndrome (PCOS) patients to elucidate PCOS-related pathways. METHODS Twenty women and their affected available relatives diagnosed with PCOS according to Rotterdam criteria were recruited. Whole-exome sequencing on germ-line DNA from 31 PCOS probands and their affected relatives was performed. Whole-exome sequencing data were further evaluated by pathway and chemogenomics analyses. In-slico analysis of candidate variants were done by VarCards for functional predictions and VarSite for impact on three-dimensional (3D) structures in the candidate proteins. RESULTS Two heterozygous rare FBN3 missense variants in three patients, and one FN1 missense variant in one patient from three different PCOS families were identified. CONCLUSION We identified three novel FBN3 and FN1 variants for the first time in the literature and linked with PCOS. Further functional studies may identify causality of these newly discovered PCOS-related variants, and their role yet remains to be investigated. Our findings may improve our understanding of the biological pathways affected and identify new drug targets.
Collapse
Affiliation(s)
- Cengiz Karakaya
- Department of Medical Biochemistry, Gazi University School of Medicine, Ankara, Turkey.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aylin Pelin Çil
- American Hospital Women's Health and Assisted Reproductive Center Guzelbahce Sok, İstanbul, Turkey
| | - Kaya Bilguvar
- Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut, USA.,Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Medical Genetics, Acibadem University School of Medicine, Istanbul, Turkey
| | - Tunahan Çakir
- Department of Bioengineering, Gebze Technical University, Gebze, Turkey
| | - Mete Hakan Karalok
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Recep Onur Karabacak
- Department of Obstetrics and Gynecology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ahmet Okay Caglayan
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Medical Genetics, School of Medicine, Dokuz Eylul University, Izmir, Turkey.,Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
6
|
Han Y, Gao G, Li S, Xiao N, Zhang Y, Luo H. Development of an optimal protocol for isolation and purification of human granulosa cells in patients with different ovarian reserves. Exp Ther Med 2021; 22:938. [PMID: 34335887 DOI: 10.3892/etm.2021.10370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
The aim of the current study was to develop an accurate and reproducible method for isolation of granulosa cells (GCs) in patients with different ovarian reserves. The cells of healthy individuals and patients with polycystic ovary syndrome (PCOS) were isolated using a modified two-step Percoll density gradient centrifugation. The cells of patients with poor ovarian response (POR) were isolated using a one-step method suitable for samples containing few cells. Cells extracted using these purification techniques were compared regarding cell yield, viability and purity using immunocytochemistry, flow cytometry, Cell Counting Kit-8, western blotting and RNA integrity analysis. The purity and activity of the cells in the POR group were comparable with those in the healthy and PCOS groups and no statistically significant differences were identified. Furthermore, isolated cells analyzed for RNA integrity indicated good quality RNA and presented an RNA integrity number of 8-10, demonstrating that the technique enabled the isolation of GCs from different types of patients. Thus, a reliable and reproducible technique for the isolation of pure GCs with high yield is described in the present study. This protocol provides an efficient method targeted to patients with different ovarian reserve functions that enables the preparation of GCs for evaluating their molecular functions.
Collapse
Affiliation(s)
- Ying Han
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| | - Ge Gao
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| | - Shuang Li
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| | - Nan Xiao
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| | - Yinfeng Zhang
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| | - Haining Luo
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, P.R. China
| |
Collapse
|
7
|
Wang Y, Cao J, Liu W, Zhang J, Wang Z, Zhang Y, Hou L, Chen S, Hao P, Zhang L, Zhuang M, Yu Y, Li D, Fan G. Protein tyrosine phosphatase receptor type R (PTPRR) antagonizes the Wnt signaling pathway in ovarian cancer by dephosphorylating and inactivating β-catenin. J Biol Chem 2019; 294:18306-18323. [PMID: 31653698 DOI: 10.1074/jbc.ra119.010348] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/15/2019] [Indexed: 11/06/2022] Open
Abstract
Despite a lack of mutations, accumulating evidence supports an important role for the Wnt/β-catenin pathway in ovarian tumorigenesis. However, the molecular mechanism that contributes to the aberrant activation of the Wnt signaling cascade in ovarian cancer has not been fully elucidated. Here, we found that protein tyrosine phosphatase receptor type R (PTPRR) suppressed the activation of the Wnt/β-catenin pathway in ovarian cancer. We performed an shRNA-based biochemical screen, which identified PTPRR as being responsible for tyrosine dephosphorylation of β-catenin on Tyr-142, a key site controlling the transcriptional activity of β-catenin. Of note, PTPRR was down-regulated in ovarian cancers, and ectopic PTPRR re-expression delayed ovarian cancer cell growth both in vitro and in vivo Using a proximity-based tagging system and RNA-Seq analysis, we identified a signaling nexus that includes PTPRR, α-catenin, β-catenin, E-cadherin, and AT-rich interaction domain 3C (ARID3C) in ovarian cancer. Immunohistochemistry staining of human samples further suggested that PTPRR expression is inversely correlated with disease prognosis. Collectively, our findings indicate that PTPRR functions as a tumor suppressor in ovarian cancer by dephosphorylating and inactivating β-catenin. These results suggest that PTPRR expression might have utility as a prognostic marker for predicting overall survival.
Collapse
Affiliation(s)
- Yuetong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Weiwei Liu
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiali Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zuo Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yiqun Zhang
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Linjun Hou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shengmiao Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Liye Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yang Yu
- Institute of Biophysics, Key Laboratory of RNA Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Dake Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
8
|
Hassani F, Oryan S, Eftekhari-Yazdi P, Bazrgar M, Moini A, Nasiri N, Sharifi-Zarchi A. Downregulation of Extracellular Matrix and Cell Adhesion Molecules in Cumulus Cells of Infertile Polycystic Ovary Syndrome Women with and without Insulin Resistance. CELL JOURNAL 2018; 21:35-42. [PMID: 30507086 PMCID: PMC6275428 DOI: 10.22074/cellj.2019.5576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/27/2018] [Indexed: 11/04/2022]
Abstract
Objective The extracellular matrix (ECM) of the cumulus oocyte complex (COC) is composed of several molecules that have different roles during follicle development. This study aims to explore gene expression profiles for ECM and cell adhesion molecules in the cumulus cells of polycystic ovary syndrome (PCOS) patients based on their insulin sensitivity following controlled ovarian stimulation (COS). Materials and Methods In this prospective case-control study enrolled 23 women less than 36 years of age who participated in an intracytoplasmic sperm injection (ICSI) program. Patients were subdivided into 3 groups: control (n=8, fertile women with male infertility history), insulin resistant (IR) PCOS (n=7), and insulin sensitive (IS) PCOS (n=8). We compared 84 ECM component and adhesion molecule gene expressions by quantitative real-time polymerase chain reaction array (qPCR-array) among the groups. Results We noted that 21 of the 84 studied genes differentially expressed among the groups, from which 18 of these genes downregulated. Overall, comparison of PCOS cases with controls showed downregulation of extracellular matrix protein 1 (ECM1); catenin (cadherin-associated protein), alpha 1 (CTNNA1); integrin, alpha 5 (ITGA5); laminin, alpha 3 (LAMA3); laminin, beta 1 (LAMB1); fibronectin 1 (FN1); and integrin, alpha 7 (ITGA7). In the IS group, there was upregulation of ADAM metallopeptidase with thrombospondin type 1 motif, 8 (ADAMTS8) and neural cell adhesion molecule 1 (NCAM1) compared with the controls (P<0.05). Conclusion Downregulation of ECM and cell adhesion molecules seem to be related to PCOS. Gene expression profile alterations in cumulus cells from both the IS and IR groups of PCOS patients seems to be involved in the composition and regulation of ECM during the ovulation process. This study highlights the association of ECM gene alteration as a viewpoint for additional understanding of the etiology of PCOS.
Collapse
Affiliation(s)
- Fatemeh Hassani
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.Electronic Address:
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Electronic Address:
| | - Masood Bazrgar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nasiri
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ali Sharifi-Zarchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Huang X, Pan J, Wu B, Teng X. Construction and analysis of a lncRNA (PWRN2)-mediated ceRNA network reveal its potential roles in oocyte nuclear maturation of patients with PCOS. Reprod Biol Endocrinol 2018; 16:73. [PMID: 30075721 PMCID: PMC6091030 DOI: 10.1186/s12958-018-0392-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/25/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in women. An lncRNA, namely, Prader-Willi region nonprotein coding RNA 2 (PWRN2), was up-regulated in the cumulus cells of patients with PCOS. However, the molecular mechanism of PWRN2 in PCOS remains largely unknown. METHODS In this study, the expression levels of PWRN2 were tested in cumulus cells through qRT-PCR analysis to confirm its potential roles in oocyte nuclear maturation of PCOS. A PWRN2-mediated ceRNA network was constructed based on three microarray datasets to investigate the molecular mechanism of PWRN2 in oocyte development of patients with PCOS. The direct interactions of the candidate genes of the ceRNA network were also demonstrated by dual-luciferase reporter assay. RESULTS PWRN2 was found to be associated with oocyte nuclear maturation in patients with PCOS in contrast to that in normal patients. Based on the microarray data, 176 lncRNAs (118 up-regulated and 58 down-regulated) and 131 mRNAs (84 up-regulated and 47 down-regulated) were identified to be regulated by PWRN2. A PWRN2-miR-92b-3p-TMEM120B ceRNA network was constructed based on results of analysis of the combined three microarray datasets (lncRNA+mRNA microarray in KGN/shPWRN2 in this study, miRNAs microarray and lncRNA+mRNA microarray in PCOS cumulus cells reported in previous studies). The coexpression characteristics of the genes (PWRN2, miR-92b-3p and TMEM120B) were detected in the cumulus cells of cumulus-oocyte complexes at different nuclear maturity stages in PCOS. These results are in accordance with the ceRNA hypothesis. Moreover, luciferase activity assay revealed that miR-92b-3p directly binds to PWRN2 and targets TMEM120B. CONCLUSIONS PWNR2 plays important roles in oocyte nuclear maturation in PCOS by functioning as a ceRNA to reduce the availability of miR-92b-3p for TMEM120B target binding during oocyte maturation in PCOS. Our findings would provide new information and clarify abnormal oocyte development in PCOS.
Collapse
Affiliation(s)
- Xin Huang
- 0000000123704535grid.24516.34Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 2699 Gaoke Road West, Shanghai, 200001 People’s Republic of China
| | - Jiaping Pan
- 0000000123704535grid.24516.34Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 2699 Gaoke Road West, Shanghai, 200001 People’s Republic of China
| | - Bi Wu
- 0000000123704535grid.24516.34Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 2699 Gaoke Road West, Shanghai, 200001 People’s Republic of China
| | - Xiaoming Teng
- 0000000123704535grid.24516.34Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 2699 Gaoke Road West, Shanghai, 200001 People’s Republic of China
| |
Collapse
|
10
|
Shi M, Cheng J, He Y, Jiang Z, Bodinga BM, Liu B, Chen H, Li Q. Effect of FH535 on in vitro
maturation of porcine oocytes by inhibiting WNT signaling pathway. Anim Sci J 2017; 89:631-639. [DOI: 10.1111/asj.12982] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/26/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Meihong Shi
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Jianyong Cheng
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Yamei He
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Zhongliang Jiang
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Bello M. Bodinga
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Boyang Liu
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Huali Chen
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Qingwang Li
- College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
- College of Environment and Chemistry Engineering; Yanshan University; Qinhuangdao Hebei China
| |
Collapse
|
11
|
Huang X, Liu C, Hao C, Tang Q, Liu R, Lin S, Zhang L, Yan W. Identification of altered microRNAs and mRNAs in the cumulus cells of PCOS patients: miRNA-509-3p promotes oestradiol secretion by targeting MAP3K8. Reproduction 2016; 151:643-55. [PMID: 27001999 DOI: 10.1530/rep-16-0071] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in women and is characterised by polycystic ovaries, hyperandrogenism and chronic anovulation. Although the clinical and biochemical signs of PCOS are typically heterogeneous, abnormal folliculogenesis is considered a common characteristic of PCOS. Our aim is to identify the altered miRNA and mRNA expression profiles in the cumulus cells of PCOS patients to investigate their molecular function in the aetiology and pathophysiology of PCOS. In this study, the miRNA expression profiles of the cumulus cell samples isolated from five PCOS and five control patients were determined by an miRNA microarray. At the same time, the altered mRNA profiles of the same cumulus cell samples were also identified by a cDNA microarray. From the microarray data, 17 miRNAs and 1263 mRNAs showed significantly different expression in the PCOS cumulus cells. The differentially expressed miRNA-509-3p and its potential target gene (MAP3K8) were identified from the miRNA and mRNA microarrays respectively. The expression of miRNA-509-3p was up-regulated and MAP3K8 was down-regulated in the PCOS cumulus cells. The direct interaction between miRNA-509-3p and MAP3K8 was confirmed by a luciferase activity assay in KGN cells. In addition, miRNA-509-3p mimics or inhibitor transfection tests in KGN cells further confirmed that miRNA-509-3p improved oestradiol (E2) secretion by inhibiting the expression of MAP3K8 These results help to characterise the pathogenesis of anovulation in PCOS, especially the regulation of E2 production.
Collapse
Affiliation(s)
- Xin Huang
- Reproductive Medicine CentreAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Chang Liu
- Shandong University School of MedicineJinan, Shandong, People's Republic of China
| | - Cuifang Hao
- Reproductive Medicine CentreAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Qianqing Tang
- Department of LaboratoryAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Riming Liu
- Department of LaboratoryAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Shaoxia Lin
- Department of LaboratoryAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Luping Zhang
- Reproductive Medicine CentreAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| | - Wei Yan
- Reproductive Medicine CentreAffiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, Yantai, Shandong, People's Republic of China
| |
Collapse
|
12
|
Huang X, Hao C, Bao H, Wang M, Dai H. Aberrant expression of long noncoding RNAs in cumulus cells isolated from PCOS patients. J Assist Reprod Genet 2015; 33:111-21. [PMID: 26650608 DOI: 10.1007/s10815-015-0630-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/29/2015] [Indexed: 01/14/2023] Open
Abstract
PURPOSE To describe the long noncoding RNA (lncRNA) profiles in cumulus cells isolated from polycystic ovary syndrome (PCOS) patients by employing a microarray and in-depth bioinformatics analysis. This information will help us understand the occurrence and development of PCOS. METHODS In this study, we used a microarray to describe lncRNA profiles in cumulus cells isolated from ten patients (five PCOS and five normal women). Several differentially expressed lncRNAs were chosen to validate the microarray results by quantitative RT-PCR (qRT-PCR). Then, the differentially expressed lncRNAs were classified into three subgroups (HOX loci lncRNA, enhancer-like lncRNA, and lincRNA) to deduce their potential features. Furthermore, a lncRNA/mRNA co-expression network was constructed by using the Cytoscape software (V2.8.3, http://www.cytoscape.org/ ). RESULTS We observed that 623 lncRNAs and 260 messenger RNAs (mRNAs) were significantly up- or down-regulated (≥2-fold change), and these differences could be used to discriminate cumulus cells of PCOS from those of normal patients. Five differentially expressed lncRNAs (XLOC_011402, ENST00000454271, ENST00000433673, ENST00000450294, and ENST00000432431) were selected to validate the microarray results using quantitative RT-PCR (qRT-PCR). The qRT-PCR results were consistent with the microarray data. Further analysis indicated that many differentially expressed lncRNAs were transcribed from chromosome 2 and may act as enhancers to regulate their neighboring protein-coding genes. Forty-three lncRNAs and 29 mRNAs were used to construct the coding-non-coding gene co-expression network. Most pairs positively correlated, and one mRNA correlated with one or more lncRNAs. CONCLUSIONS Our study is the first to determine genome-wide lncRNA expression patterns in cumulus cells isolated from PCOS patients by microarray. The results show that clusters of lncRNAs were aberrantly expressed in cumulus cells of PCOS patients compared with those of normal women, which revealed that lncRNAs differentially expressed in PCOS and normal women may contribute to the occurrence of PCOS and affect oocyte development.
Collapse
Affiliation(s)
- Xin Huang
- Reproductive Medicine Centre, Affiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, 20 Yuhuangding Road East, Yantai, Shandong, 264000, People's Republic of China.
| | - Cuifang Hao
- Reproductive Medicine Centre, Affiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, 20 Yuhuangding Road East, Yantai, Shandong, 264000, People's Republic of China.
| | - Hongchu Bao
- Reproductive Medicine Centre, Affiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, 20 Yuhuangding Road East, Yantai, Shandong, 264000, People's Republic of China.
| | - Meimei Wang
- Reproductive Medicine Centre, Affiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, 20 Yuhuangding Road East, Yantai, Shandong, 264000, People's Republic of China.
| | - Huangguan Dai
- Reproductive Medicine Centre, Affiliated Hospital of Qingdao Medical University, Yuhuangding Hospital of Yantai, 20 Yuhuangding Road East, Yantai, Shandong, 264000, People's Republic of China.
| |
Collapse
|
13
|
Schmidt J, Weijdegard B, Mikkelsen AL, Lindenberg S, Nilsson L, Brannstrom M. Differential expression of inflammation-related genes in the ovarian stroma and granulosa cells of PCOS women. Mol Hum Reprod 2013; 20:49-58. [DOI: 10.1093/molehr/gat051] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
14
|
Takahashi N, Tarumi W, Ishizuka B. Acute reproductive toxicity of 3,3′-iminodipropionitrile in female rats. Reprod Toxicol 2012; 33:27-34. [DOI: 10.1016/j.reprotox.2011.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 10/03/2011] [Accepted: 10/24/2011] [Indexed: 11/28/2022]
|
15
|
Microarray analysis in gynaecology and its findings: a systematic review. Reprod Biomed Online 2011; 22:569-82. [PMID: 21507720 DOI: 10.1016/j.rbmo.2011.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 02/24/2011] [Accepted: 03/10/2011] [Indexed: 11/21/2022]
Abstract
Microarray technology is a promising method for investigating gynaecological benign pathology. This systematic review examined various parameters of the design of these studies, the methods used and the gene outcome in these diseases. Electronic searches were performed in Medline (up to April 2009). An overall representation of important genes for each disease detected was performed. The results showed genes were up-regulated or down-regulated. However, studies suffer from several flaws in their design, the sample size employed and the reporting method. In conclusion, a significant amount of work has been performed on benign gynaecological diseases using microarray technology. New trial designs need to be employed that incorporate microarray reporting standards. New research directions should evolve based on these results.
Collapse
|
16
|
Papachroni KK, Piperi C, Levidou G, Korkolopoulou P, Pawelczyk L, Diamanti-Kandarakis E, Papavassiliou AG. Lysyl oxidase interacts with AGE signalling to modulate collagen synthesis in polycystic ovarian tissue. J Cell Mol Med 2011; 14:2460-9. [PMID: 19583806 PMCID: PMC3823163 DOI: 10.1111/j.1582-4934.2009.00841.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Connective tissue components – collagen types I, III and IV – surrounding the ovarian follicles undergo drastic changes during ovulation. Abnormal collagen synthesis and increased volume and density of ovarian stroma characterize the polycystic ovary syndrome (PCOS). During the ovulatory process, collagen synthesis is regulated by prolyl hydroxylase and lysyl oxidase (LOX) activity in ovarian follicles. LOX catalyzes collagen and elastin cross-linking and plays essential role in coordinating the control of ovarian extracellular matrix (ECM) during follicular development. We have recently shown accumulation of advanced glycation end products (AGEs), molecules that stimulate ECM production and abnormal collagen cross-linking, in ovarian tissue. However, the possible link between LOX and AGEs-induced signalling in collagen production and stroma formation in ovarian tissue from PCOS remains elusive. The present study investigates the hypothesis of AGE signalling pathway interaction with LOX gene activity in polycystic ovarian (PCO) tissue. We show an increased distribution and co-localization of LOX, collagen type IV and AGE molecules in the PCO tissue compared to control, as well as augmented expression of AGE signalling mediators/effectors, phospho(p)-ERK, phospho(p)-c-Jun and nuclear factor κB (NF-κB) in pathological tissue. Moreover, we demonstrate binding of AGE-induced transcription factors, NF-κB and activator protein-1 (AP-1) on LOX promoter, indicating a possible involvement of AGEs in LOX gene regulation, which may account for the documented increase in LOX mRNA and protein levels compared to control. These findings suggest that deposition of excess collagen in PCO tissue that induces cystogenesis may, in part, be due to AGE-mediated stimulation of LOX activity.
Collapse
Affiliation(s)
- Katerina K Papachroni
- Department of Biological Chemistry, Medical School, University of Athens, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
17
|
Kitaya K, Yamada H. Pathophysiological roles of chemokines in human reproduction: an overview. Am J Reprod Immunol 2010; 65:449-59. [PMID: 21087337 DOI: 10.1111/j.1600-0897.2010.00928.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a group of small cytokines that have an ability to induce leukocyte migration. Chemokines exert their functions by binding and activating specific G protein-coupled receptors. Studies have unveiled pleiotropic bioactivities of chemokines in various phenomena ranging from immunomodulation, embryogenesis, and homeostasis to pathogenesis. In the mammalian reproductive system, chemokines unexceptionally serve in multimodal events that are closely associated with establishment, maintenance, and deterioration of fecundity. The aim of this review is to update the knowledge on chemokines in male and female genital organs, with a focus on their potential pathophysiological roles in human reproduction.
Collapse
Affiliation(s)
- Kotaro Kitaya
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan.
| | | |
Collapse
|
18
|
Assou S, Boumela I, Haouzi D, Anahory T, Dechaud H, De Vos J, Hamamah S. Dynamic changes in gene expression during human early embryo development: from fundamental aspects to clinical applications. Hum Reprod Update 2010; 17:272-90. [PMID: 20716614 DOI: 10.1093/humupd/dmq036] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The first week of human embryonic development comprises a series of events that change highly specialized germ cells into undifferentiated human embryonic stem cells (hESCs) that display an extraordinarily broad developmental potential. The understanding of these events is crucial to the improvement of the success rate of in vitro fertilization. With the emergence of new technologies such as Omics, the gene expression profiling of human oocytes, embryos and hESCs has been performed and generated a flood of data related to the molecular signature of early embryo development. METHODS In order to understand the complex genetic network that controls the first week of embryo development, we performed a systematic review and study of this issue. We performed a literature search using PubMed and EMBASE to identify all relevant studies published as original articles in English up to March 2010 (n = 165). We also analyzed the transcriptome of human oocytes, embryos and hESCs. RESULTS Distinct sets of genes were revealed by comparing the expression profiles of oocytes, embryos on Day 3 and hESCs, which are associated with totipotency, pluripotency and reprogramming properties, respectively. Known components of two signaling pathways (WNT and transforming growth factor-β) were linked to oocyte maturation and early embryonic development. CONCLUSIONS Omics analysis provides tools for understanding the molecular mechanisms and signaling pathways controlling early embryonic development. Furthermore, we discuss the clinical relevance of using a non-invasive molecular approach to embryo selection for the single-embryo transfer program.
Collapse
Affiliation(s)
- Said Assou
- CHU Montpellier, Institute for Research in Biotherapy, Hôpital Saint-Eloi, INSERM U847, Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Liu JJ, Ma X, Cai LB, Cui YG, Liu JY. Downregulation of both gene expression and activity of Hsp27 improved maturation of mouse oocyte in vitro. Reprod Biol Endocrinol 2010; 8:47. [PMID: 20465849 PMCID: PMC2890611 DOI: 10.1186/1477-7827-8-47] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 05/14/2010] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Heat shock protein 27 (Hsp27), a member of the small heat shock protein family, is an apoptosis regulator. Our previous proteomic study showed that Hsp27 mainly expressed in human oocyte, and that Hsp27 expression was downregulated in the ovaries derived from women with the polycystic ovary syndrome (PCOS), a well known endocrinal disorder with abnormal apoptotic activity and folliculogenesis. However, the exact effects of Hsp27 downregulation on oocyte development have not yet been clarified. METHODS The expression of Hsp27 gene was downregulated in the mouse oocytes cultured in vitro using siRNA adenovirus infection, while the activity of Hsp27 was decreased by microinjection of polyclonal Hsp27 antibody into the cytoplasm of germinal vesicle (GV) oocytes. Oocyte maturation rate was evaluated by morphological observation. Early stage of apoptosis was determined using Annexin-V staining analysis and some critical apoptotic factors and cytokines were also monitored at both mRNA level by real time RT-PCR and protein expression level by immunofluorescence and western blot. RESULTS Hsp27 expressed at high level in maturing oocytes. Infection with AdshHsp27, and microinjection of Hsp27 antibody into GV oocytes, resulted in the improved oocyte development and maturation. Germinal vesicle breakdown (GVBD) rates were significantly increased in two AdshHsp27-treated groups (88.7%, 86.0%) and Hsp27 antibody-injected group (77.0%) when compared with control (76.2% in AdGFP, 64.4% in IgG-injected), respectively. In addition, the rates of metaphase II (MII) development in two AdshHsp27-treated groups (73.8%, 76.4%) and Hsp27 antibody-injected group (67.3%) were higher than that in the controls (59.6% in AdGFP, 55.1% in IgG-injected). We also found that the rates of early stage of apoptosis in Hsp27 downregulated groups (46.5% and 45.6%) were higher than that in control group (34.1%) after 8 h of IVM. Similarly, downregulation of Hsp27 caused a significantly enhanced the expression of apoptotic factors (caspase 8, caspase 3) and cytokines (bmp 15 and gdf 9). CONCLUSIONS Downregulation of Hsp27 improved the maturation of mouse oocytes, while increased early stage of apoptosis in oocytes by inducing the activation of extrinsic, caspase 8-mediated pathway.
Collapse
Affiliation(s)
- Jin-Juan Liu
- Department of life science and technology, China Pharmaceutical University, Nanjing 210038, China
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Xiang Ma
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Ling-Bo Cai
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yu-Gui Cui
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Jia-Yin Liu
- Department of life science and technology, China Pharmaceutical University, Nanjing 210038, China
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
20
|
Mahmoudi T, Gourabi H, Ashrafi M, Yazdi RS, Ezabadi Z. Calciotropic hormones, insulin resistance, and the polycystic ovary syndrome. Fertil Steril 2010; 93:1208-14. [DOI: 10.1016/j.fertnstert.2008.11.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 11/24/2008] [Accepted: 11/26/2008] [Indexed: 10/21/2022]
|
21
|
Boyer A, Goff AK, Boerboom D. WNT signaling in ovarian follicle biology and tumorigenesis. Trends Endocrinol Metab 2010; 21:25-32. [PMID: 19875303 DOI: 10.1016/j.tem.2009.08.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 08/27/2009] [Accepted: 08/28/2009] [Indexed: 01/07/2023]
Abstract
The WNTS are an expansive family of glycoprotein signaling molecules known mostly for the roles they play in embryonic development. WNT signaling first caught the attention of ovarian biologists when it was reported that the inactivation of Wnt4 in mice results in partial female-to-male sex reversal and oocyte depletion. More recently, studies using loss- and gain-of-function transgenic mouse models demonstrated the requirement for Wnt4, Fzd4 and Ctnnb1, components of the WNT pathway, for normal folliculogenesis, luteogenesis and steroidogenesis, and showed that dysregulated WNT signaling can cause granulosa cell tumor development. This review covers our current knowledge of WNT signaling in ovarian follicles, highlighting both the great promise and the many unresolved questions of this emerging field of research.
Collapse
Affiliation(s)
- Alexandre Boyer
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | | | | |
Collapse
|
22
|
Mohamed-Hussein ZA, Harun S. Construction of a polycystic ovarian syndrome (PCOS) pathway based on the interactions of PCOS-related proteins retrieved from bibliomic data. Theor Biol Med Model 2009; 6:18. [PMID: 19723303 PMCID: PMC2743649 DOI: 10.1186/1742-4682-6-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Accepted: 09/01/2009] [Indexed: 12/15/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex but frequently occurring endocrine abnormality. PCOS has become one of the leading causes of oligo-ovulatory infertility among premenopausal women. The definition of PCOS remains unclear because of the heterogeneity of this abnormality, but it is associated with insulin resistance, hyperandrogenism, obesity and dyslipidaemia. The main purpose of this study was to identify possible candidate genes involved in PCOS. Several genomic approaches, including linkage analysis and microarray analysis, have been used to look for candidate PCOS genes. To obtain a clearer view of the mechanism of PCOS, we have compiled data from microarray analyses. An extensive literature search identified seven published microarray analyses that utilized PCOS samples. These were published between the year of 2003 and 2007 and included analyses of ovary tissues as well as whole ovaries and theca cells. Although somewhat different methods were used, all the studies employed cDNA microarrays to compare the gene expression patterns of PCOS patients with those of healthy controls. These analyses identified more than a thousand genes whose expression was altered in PCOS patients. Most of the genes were found to be involved in gene and protein expression, cell signaling and metabolism. We have classified all of the 1081 identified genes as coding for either known or unknown proteins. Cytoscape 2.6.1 was used to build a network of protein and then to analyze it. This protein network consists of 504 protein nodes and 1408 interactions among those proteins. One hypothetical protein in the PCOS network was postulated to be involved in the cell cycle. BiNGO was used to identify the three main ontologies in the protein network: molecular functions, biological processes and cellular components. This gene ontology analysis identified a number of ontologies and genes likely to be involved in the complex mechanism of PCOS. These include the insulin receptor signaling pathway, steroid biosynthesis, and the regulation of gonadotropin secretion among others.
Collapse
Affiliation(s)
- Zeti-Azura Mohamed-Hussein
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, UKM Bangi, Selangor, Malaysia.
| | | |
Collapse
|
23
|
Kenigsberg S, Bentov Y, Chalifa-Caspi V, Potashnik G, Ofir R, Birk OS. Gene expression microarray profiles of cumulus cells in lean and overweight-obese polycystic ovary syndrome patients. Mol Hum Reprod 2009; 15:89-103. [PMID: 19141487 DOI: 10.1093/molehr/gan082] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The aim of this work was to study gene expression patterns of cultured cumulus cells from lean and overweight-obese polycystic ovary syndrome (PCOS) patients using genome-wide oligonucleotide microarray. The study included 25 patients undergoing in vitro fertilization and intra-cytoplasmic sperm injection: 12 diagnosed with PCOS and 13 matching controls. Each of the groups was subdivided into lean (body mass index (BMI) < 24) and overweight (BMI > 27) subgroups. The following comparisons of gene expression data were made: lean PCOS versus lean controls, lean PCOS versus overweight PCOS, all PCOS versus all controls, overweight PCOS versus overweight controls, overweight controls versus lean controls and all overweight versus all lean. The largest number of differentially expressed genes (DEGs), with fold change (FC) |FC| >or= 1.5 and P-value < 0.01, was found in the lean PCOS versus lean controls comparison (487) with most of these genes being down-regulated in PCOS. The second largest group of DEGs originated from the comparison of lean PCOS versus overweight PCOS (305). The other comparisons resulted in a much smaller number of DEGs (174, 109, 125 and 12, respectively). In the comparison of lean PCOS with lean controls, most DEGs were transcription factors and components of the extracellular matrix and two pathways, Wnt/beta-catenin and mitogen-activated protein kinase. When comparing overweight PCOS with overweight controls, most DEGs were of pathways related to insulin signaling, metabolism and energy production. The finding of unique gene expression patterns in cumulus cells from the two PCOS subtypes is in agreement with other studies that have found the two to be separate entities with potentially different pathophysiologies.
Collapse
Affiliation(s)
- Shlomit Kenigsberg
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| | | | | | | | | | | |
Collapse
|
24
|
Escobar-Morreale HF, Insenser M, Cortón M, Millán JLS, Peral B. Proteomics and genomics: A hypothesis-free approach to the study of the role of visceral adiposity in the pathogenesis of the polycystic ovary syndrome. Proteomics Clin Appl 2008; 2:444-55. [DOI: 10.1002/prca.200780031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
25
|
Polycystic ovarian syndrome: pathophysiology, molecular aspects and clinical implications. Expert Rev Mol Med 2008; 10:e3. [PMID: 18230193 DOI: 10.1017/s1462399408000598] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is universally recognised as the commonest endocrinopathy of women. The definition and the aetiological hypotheses of PCOS are continuously evolving to accommodate expanding knowledge on the syndrome, which is now known to be more complex than purely a reproductive disorder. Increased androgen synthesis, disrupted folliculogenesis and insulin resistance lie at the pathophysiological core of PCOS. An intriguing concept involves the perpetuation of a vicious circle with endocrine/reproductive and metabolic components. An unfavourable metabolic environment may unmask genetic traits of ovarian dysfunction, and the unfolding endocrine derangement could further aggravate the metabolic disarray. This article reviews the molecular mechanisms known to underlie the ovarian and metabolic abnormalities characterising PCOS. The putative interdependence between reproductive and metabolic aspects of PCOS, and therapeutic implications for the management of PCOS, are also discussed.
Collapse
|
26
|
Wood JR, Dumesic DA, Abbott DH, Strauss JF. Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J Clin Endocrinol Metab 2007; 92:705-13. [PMID: 17148555 DOI: 10.1210/jc.2006-2123] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility, is characterized by increased ovarian androgen production and arrested follicle development and is frequently associated with insulin resistance. These PCOS phenotypes are associated with exaggerated ovarian responsiveness to FSH and increased pregnancy loss. OBJECTIVE The objective of this study was to examine whether the perturbations in follicle growth and the intrafollicular environment affect gene expression and ultimately development of the PCOS oocyte. DESIGN Oocyte cDNA was subjected to microarray and PCR analysis. SETTING This study was conducted at a university laboratory. PATIENTS The study comprised 10 normal ovulatory women and nine women with PCOS. INTERVENTION The intervention was GnRH analog/recombinant human FSH therapy for in vitro fertilization. MAIN OUTCOME MEASURE The main outcome measure was mRNA abundance of oocyte-expressed genes. RESULTS Cluster analysis revealed differences in global gene expression profiles between normal and PCOS oocytes. Of the 8123 transcripts expressed in the oocytes, 374 genes showed significant differences in mRNA abundance in PCOS oocytes. Annotation of the data demonstrated that a subset of these genes was associated with chromosome alignment and segregation during mitosis and/or meiosis. Furthermore, 68 of the differentially expressed genes contained putative androgen receptor and/or peroxisome proliferating receptor gamma binding sites. CONCLUSIONS These analyses demonstrated that normal and PCOS oocytes that are morphologically indistinguishable and of high quality exhibit different gene expression profiles. Promoter analysis suggests that androgens and other activators of nuclear receptors may play a role in differential gene expression in the PCOS oocyte. Likewise, annotation of the differentially expressed genes suggests that defects in meiosis or early embryonic development may contribute to reduced developmental competency of PCOS oocytes.
Collapse
Affiliation(s)
- Jennifer R Wood
- Department of Animal Science, University of Nebraska-Lincoln, 3800 Fair Street, Lincoln, Nebraska 68583-0908, USA.
| | | | | | | |
Collapse
|