1
|
Xu X, Yin C, Dong B, Li Y, Liu S, Chen J. LncRNA XIST Protects Against Polycystic Ovary Syndrome via the Regulation of miR-212-3p/RASA1 Axis. Biochem Genet 2025; 63:1686-1698. [PMID: 38609670 DOI: 10.1007/s10528-024-10777-7] [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: 10/08/2023] [Accepted: 03/07/2024] [Indexed: 04/14/2024]
Abstract
The polycystic ovary syndrome (PCOS), a common endocrine disorder, is mainly related to infertility. Moreover, it is characterized by promoted androgen, suppressed ovulation and insulin resistance. Long non-coding RNA X inactive specific transcript (lncRNA XIST), known as an oncogene or a cancer inhabited factor, is involved in several disease. However, the diagnostic mechanisms of lncRNA XIST in PCOS have not been clarified. Our study aimed to explain whether lncRNA XIST regulates KGN cells proliferation and apoptosis via microRNA (miR)-212-3p/RASA1 axis in PCOS. Levels of lncRNA XIST, miR-212-3p and RASA1 in KGN cells were detected through reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay. Fluorescence in situ Hybridization (FISH) was performed to confirm the expression of lncRNA XIST and miR-212-3p in KGN cells. StarBase and dual-luciferase reporter assay were applied for exploring the interaction between miR-212-3p and RASA1. Cell viability, apoptosis, protein expression of Bcl-2 and Bax were assessed by MTT, flow cytometry analysis, RT-qPCR and western blot, respectively. We found that lncRNA XIST was low-expressed, miR-212-3p was over-expressed, and RASA1 was dramatically down-regulated in KGN cells. LncRNA XIST negatively regulated miR-212-3p expression in KGN cells. MiR-212-3p interacted with RASA1 and negatively regulated RASA1 levels in KGN cells. Up-regulation of lncRNA XIST signally decreased cells viability, stimulated more apoptotic cells, enhanced Bax expression, and depressed Bcl-2 level in KGN cells. However, these observations were abolished after miR-212-3p mimic treatment. Furthermore, miR-212-3p inhibitor significantly inhibited cell proliferation, enhanced more apoptotic cells, increased Bax expression, and decreased Bcl-2 level in KGN cells, and these effects were eliminated by RASA1-siRNA transfection. Our observations revealed that lncRNA XIST protects against PCOS through regulating miR-212-3p/RASA1 axis, suggesting that lncRNA XIST may be a promising therapeutic target for PCOS therapy.
Collapse
Affiliation(s)
- Xiaomeng Xu
- Gynecology Department 2, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Qiqihar, 161000, China.
| | - Cheng Yin
- Obstetrics Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, China
| | - Bing Dong
- Gynecology Department 2, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Qiqihar, 161000, China
| | - Yuewen Li
- Gynecology Department 2, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Qiqihar, 161000, China
| | - Shi Liu
- Central Laboratory, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, China
| | - Jun Chen
- Gynecology Department 2, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Qiqihar, 161000, China
| |
Collapse
|
2
|
Fu LW, Gao Z, Zhang N, Yang N, Long HY, Kong LY, Li XY. Traditional Chinese medicine formulae: A complementary method for the treatment of polycystic ovary syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117698. [PMID: 38171464 DOI: 10.1016/j.jep.2023.117698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/04/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polycystic ovary syndrome (PCOS) is a prevalent female endocrine condition that significantly affects women of all age groups and is characterized by metabolic dysfunction. The efficacy of existing pharmaceutical interventions for the treatment of PCOS remains inadequate. With a rich history and cultural significance spanning thousands of years, Traditional Chinese Medicine (TCM) is extensively employed for treating a variety of ailments and can serve as a supplementary therapy for managing PCOS. Multiple clinical observations and laboratory tests have unequivocally demonstrated the substantial effectiveness and safety of TCM formulae in treating PCOS, and further investigations are currently in progress. AIM OF THE STUDY To summarize the TCM formulae commonly employed in the clinical management of PCOS, examine their therapeutic benefits, investigate their mechanism of action, active constituents, and establish the correlation between efficacy, mechanism of action, and active constituents. MATERIALS AND METHODS We conducted a comprehensive search on PubMed, Web of Science, and China national knowledge infrastructure (CNKI) using the following keywords: "Polycystic Ovary Syndrome", "Traditional Chinese Medicine Decoctions", "Traditional Chinese Medicine formulae", "Traditional Chinese Medicine", "Clinical Observation", "Mechanism", "Treatment", "Pharmacology", and various combinations of these terms. From January 1, 2006 until October 7, 2023, (inclusive). RESULTS This paper summarized the clinical effectiveness, mechanism of action, and active components of 8 TCM formulae for the treatment of PCOS. Our research indicates that TCM formulae can potentially treat PCOS by enhancing the levels of hyperandrogenism and other endocrine hormones, decreasing insulin resistance and hyperinsulinemia, and controlling chronic low-grade inflammation, among other modes of action. In addition, we found an association between epigenetics and TCM formulae for the treatment of PCOS. CONCLUSION TCM formulae have specific advantages in the treatment of Polycystic Ovary Syndrome (PCOS). They achieve therapeutic benefits by targeting several pathways and connections, attracting considerable interest and playing a vital role in the treatment of PCOS. TCM formulae can be used as an adjunctive therapy for the treatment of PCOS.
Collapse
Affiliation(s)
- Li-Wen Fu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zu Gao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ning Zhang
- Department of Reproduction and Genetics, Shandong Province Hospital of Traditional Chinese, Affiliated Hospital, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Nan Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hui-Yan Long
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ling-Yuan Kong
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiu-Yang Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
3
|
Usman M, Li A, Wu D, Qinyan Y, Yi LX, He G, Lu H. The functional role of lncRNAs as ceRNAs in both ovarian processes and associated diseases. Noncoding RNA Res 2024; 9:165-177. [PMID: 38075201 PMCID: PMC10709095 DOI: 10.1016/j.ncrna.2023.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 04/26/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) have attracted significant scientific attention due to their central role in regulating gene expression and their profound impact on the intricate mechanisms of ovarian function. These versatile molecules exert their influence through various mechanisms, including the coordination of transcription processes, modulation of post-transcriptional events, and the shaping of epigenetic landscapes. Furthermore, lncRNAs function as competitive endogenous RNAs (ceRNAs), engaging in intricate interactions with microRNAs (miRNAs) to finely adjust the expression of target genes. The intricate lncRNA-miRNA-mRNA network serves as a crucial determinant in governing the multifaceted physiological functions of the ovaries. It holds substantial potential in unraveling the causes and progression of reproductive disorders and, importantly, in identifying new therapeutic targets and diagnostic markers for these conditions. A comprehensive comprehension of lncRNAs and their ceRNA activities within the domain of ovarian biology could potentially lead to groundbreaking advancements in clinical interventions and management strategies. This exploration of lncRNAs and their intricate involvement in the regulatory framework provides an extensive platform for deciphering the complex nature of ovarian physiology and pathology. The ongoing progress in this field, which encompasses in-depth investigations into the functional roles of specific lncRNAs, the elucidation of their complex interactions with miRNAs, and the comprehensive profiling of their expression patterns, holds the promise of making significant contributions to our understanding of ovarian biology and reproductive disorders. Ultimately, these breakthroughs will have wide-ranging translational implications, paving the way for the development of precision therapies and personalized medicine strategies to address the myriad challenges in the realm of reproductive health.
Collapse
Affiliation(s)
- Muhammad Usman
- Department of Plastic and Reconstructive Surgery, Central Hospital Affiliated to Chongqing University of Technology, Gonglian yicun No.1 street lijiatuo, Banan district, Chongqing, 400054, PR China
| | - Ai Li
- Department of Postdoctoral Research Workstation, The Seventh People's Hospital of Chongqing, Chongqing, PR China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yang Qinyan
- Department of Anesthesia, Central Hospital Affiliated to Chongqing University of Technology, Gonglian yicun No.1 street lijiatuo, Banan district, Chongqing, 400054, PR China
| | - Lin Xiao Yi
- Department of Radiology, The Chenjiaqiao Hospital of Shapingba District of Chongqing, PR China
| | - Guiqiong He
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, PR China
| | - Hong Lu
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Gonglian yicun No.1 street lijiatuo, Banan district, Chongqing, 400054, PR China
| |
Collapse
|
4
|
Yang Z, Wu S, He S, Han L, Zhou M, Yang J, Chen J, Wu G. LncRNA AOC4P impacts the differentiation of macrophages and T-lymphocyte by regulating the NF-κB pathways of KGN cells: Potential pathogenesis of polycystic ovary syndrome. Am J Reprod Immunol 2023; 90:e13776. [PMID: 37766402 DOI: 10.1111/aji.13776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a multifactorial endocrine disease, which is an important cause of female infertility worldwide. PCOS patients are in a state of chronic low-grade inflammation, and immune imbalance is considered as a potential cause of its pathogenesis. METHODS The expression of AOC4P in PCOS and normal ovarian granulosa cells (GCs) was detected by real-time quantitative PCR. KGN cells were induced by dihydrotestosterone at 500 ng/mL to construct the PCOS model. After lentivirus-infected, KGN cells were constructed with AOC4P overexpression cell lines, the proliferation and apoptosis levels of KGN cells in AOC4P and NC groups were detected. Human monocyte cell line (THP-1)-derived macrophages and peripheral blood mononuclear cells (PBMC) were co-cultured with KGN cells for 48 h, respectively, and the differentiation of macrophages and CD4+ T cells were detected by flow cytometry. RESULTS Decreased AOC4P expression was found in PCOS patients. After constructing the PCOS cell model, we observed that overexpression of AOC4P promoted KGN cell proliferation and inhibited apoptosis. After co-culture with AOC4P overexpressed KGN cells, M1 macrophages decreased, M2 macrophages increased, T helper cells type 1 (Th1)/Th2 ratio increased, and regulatory T cell (Treg) cells increased. Finally, we found that AOC4P inhibited the activation of the nuclear factor κ B (NF-κB) pathway in KGN cells. CONCLUSIONS In this study, we found that AOC4P regulated the NF-κB signaling pathway by inhibiting the phosphorylation of P65, thereby affecting the proliferation and apoptosis of GCs, altering the differentiation of macrophages and T cells, thus contributing to the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Zhe Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Shujuan Wu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Shaojing He
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Lu Han
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Mengqi Zhou
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Jiao Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| | - Gengxiang Wu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
5
|
Gong Z, Shi X, Xu W, Fang Y, Fang M, Yao M, Jiang Y, Sui H, Luo M. LncRNA PWRN2 promotes polycystic ovary syndrome progression via epigenetically reducing ATRX by recruiting LSD1. Reprod Biol 2023; 23:100782. [PMID: 37320994 DOI: 10.1016/j.repbio.2023.100782] [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: 03/07/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023]
Abstract
Long non-coding RNA has been shown to mediate the progression of polycystic ovary syndrome (PCOS). However, the role and mechanism of Prader-Willi region nonprotein coding RNA 2 (PWRN2) in PCOS progression remain unclear. In our study, Sprague-Dawley rat was injected with dehydroepiandrosterone to mimic PCOS rat models. HE staining was used to assess the number of benign granular cells, and serum insulin and hormone levels were detected by ELISA kit. The expression of PWRN2 was examined by qRT-PCR. Ovarian granulosa cells (GCs) proliferation and apoptosis were examined by CCK-8 assay and flow cytometry. The protein levels of apoptosis markers and Alpha thalassemia retardation syndrome X-linked (ATRX) were determined by western blot. The interaction between lysine-specific demethylase 1 (LSD1) and PWRN2 or ATRX was confirmed by RIP and ChIP assay. Our data showed that PWRN2 was upregulated and ATRX was downregulated in the ovarium tissues and serum of PCOS rat. PWRN2 knockdown promoted GCs proliferation and inhibited apoptosis. In the mechanism, PWRN2 inhibited ATRX transcription by binding with LSD1. In addition, downregulation of ATRX also eliminated the effect of sh-PWRN2 on GCs growth. In conclusion, our data suggested that PWRN2 might restrain GCs growth to promote PCOS progression, which was achieved by binding with LSD1 to inhibit ATRX transcription.
Collapse
Affiliation(s)
- Zhaoqing Gong
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Xinlei Shi
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Weizhen Xu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Yuan Fang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Meijia Fang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Minhua Yao
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Yu Jiang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China
| | - Hongshu Sui
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan City, Shandong Province 250000, PR China.
| | - Mingjiu Luo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province 271000, PR China.
| |
Collapse
|
6
|
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
|
7
|
Zhu S, Bao H, Zhang MC, Liu H, Wang Y, Lin C, Zhao X, Liu SL. KAZN as a diagnostic marker in ovarian cancer: a comprehensive analysis based on microarray, mRNA-sequencing, and methylation data. BMC Cancer 2022; 22:662. [PMID: 35710397 PMCID: PMC9204993 DOI: 10.1186/s12885-022-09747-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/09/2022] [Indexed: 11/29/2022] Open
Abstract
Background Ovarian cancer (OC) is among the deadliest malignancies in women and the lack of appropriate markers for early diagnosis leads to poor prognosis in most cases. Previous studies have shown that KAZN is involved in multiple biological processes during development, such as cell proliferation, differentiation, and apoptosis, so defects or aberrant expression of KAZN might cause queer cell behaviors such as malignancy. Here we evaluated the KAZN expression and methylation levels for possible use as an early diagnosis marker for OC. Methods We used data from Gene Expression Omnibus (GEO) microarrays, The Cancer Genome Atlas (TCGA), and Clinical Proteomic Tumor Analysis Consortium (CPTAC) to investigate the correlations between KAZN expression and clinical characteristics of OC by comparing methylation levels of normal and OC samples. The relationships among differentially methylated sites in the KAZN gene, corresponding KAZN mRNA expression levels and prognosis were analyzed. Results KAZN was up-regulated in ovarian epithelial tumors and the expression of KAZN was correlated with the patients’ survival time. KAZN CpG site cg17657618 was positively correlated with the expression of mRNA and the methylation levels were significantly differential between the group of stage “I and II” and the group of stage “III and IV”. This study also presents a new method to classify tumor and normal tissue in OC using DNA methylation pattern in the KAZN gene body region. Conclusions KAZN was involved in ovarian cancer pathogenesis. Our results demonstrate a new direction for ovarian cancer research and provide a potential diagnostic biomarker as well as a novel therapeutic target for clinical application. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09747-2.
Collapse
Affiliation(s)
- Songling Zhu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Hongxia Bao
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Meng-Chun Zhang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Huidi Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yao Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Caiji Lin
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China.,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xingjuan Zhao
- Physical Examination Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu-Lin Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, China. .,HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China. .,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China. .,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
8
|
Xi S, Li W, Li Z, Lin W, Chen L, Tian C, Yang Y, Ma L. Novel Hub genes co-expression network mediates dysfunction in a model of polycystic ovary syndrome. Am J Transl Res 2022; 14:1979-1990. [PMID: 35422941 PMCID: PMC8991170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/18/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND This study aimed to integrate DNA methylation, miRNA, and mRNA microarray data to construct a gene co-expression network for polycystic ovarian syndrome (PCOS). METHODS The weighted gene co-expression network analysis (WGCNA) was conducted to construct a PCOS-related co-expression network by using the GEO public datasets. We performed Gene Ontology and KEGG pathway enrichment analyses for a further exploration of gene function in networks. Finally, the dysfunction module consisting of a co-expression network was mapped to the PCOS patients and tried to provide guidance to the PCOS phenotyping. RESULTS Three modules (Midnightbule, Pink, and Red) were identified to be PCOS-related by WGCNA analysis. These module-related genes were enriched in cell response to stimulus, PI3K-Akt signaling pathway, insulin biological process, signaling pathway, and cytokine-cytokine receptor interaction biological processes. The multiple-factor network, including miRNA-lncRNA and DNA methylation-mRNA interaction, was closely associated with PCOS dysfunction. CONCLUSION Our study render a novel insight into the mechanisms and might provide candidate biomarkers and therapeutic targets for the classification of PCOS dysfunction.
Collapse
Affiliation(s)
- Sujuan Xi
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Weihao Li
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Zaiyi Li
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Wenjing Lin
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Lin Chen
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Chengzi Tian
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Yazhu Yang
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| | - Lin Ma
- The Reproductive Medical Center, The Seventh Affiliated Hospital of Sun Yat-sen University Shenzhen 518107, Guangdong, China
| |
Collapse
|
9
|
Tamaddon M, Azimzadeh M, Tavangar SM. microRNAs and long non-coding RNAs as biomarkers for polycystic ovary syndrome. J Cell Mol Med 2022; 26:654-670. [PMID: 34989136 PMCID: PMC8817139 DOI: 10.1111/jcmm.17139] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/05/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is known as the most common metabolic/endocrine disorder among women of reproductive age. Its complicated causality assessment and diagnostic emphasized the role of non‐coding regulatory RNAs as molecular biomarkers in studying, diagnosing and even as therapeutics of PCOS. This review discusses a comparative summary of research into microRNAs (miRNAs) and long non‐coding RNAs (lncRNAs) that are molecularly or statistically related to PCOS. We categorize the literature in terms of centering on either miRNAs or lncRNAs and discuss the combinatory studies and promising ideas as well. Additionally, we compare the pros and cons of the prominent research methodologies used for each of the abovementioned research themes and discuss how errors can be stopped from propagation by selecting correct methodologies for future research. Finally, it can be concluded that research into miRNAs and lncRNAs has the potential for identifying functional networks of regulation with multiple mRNAs (and hence, functional proteins). This new understanding may eventually afford clinicians to control the molecular course of the pathogenesis better. With further research, RNA (with statistical significance and present in the blood) may be used as biomarkers for the disease, and more possibilities for RNA therapy agents can be identified.
Collapse
Affiliation(s)
- Mona Tamaddon
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Azimzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Saadat N, Puttabyatappa M, Elangovan VR, Dou J, Ciarelli JN, Thompson RC, Bakulski KM, Padmanabhan V. Developmental Programming: Prenatal Testosterone Excess on Liver and Muscle Coding and Noncoding RNA in Female Sheep. Endocrinology 2022; 163:6413684. [PMID: 34718504 PMCID: PMC8667859 DOI: 10.1210/endocr/bqab225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Indexed: 11/19/2022]
Abstract
Prenatal testosterone (T)-treated female sheep manifest peripheral insulin resistance, ectopic lipid accumulation, and insulin signaling disruption in liver and muscle. This study investigated transcriptional changes and transcriptome signature of prenatal T excess-induced hepatic and muscle-specific metabolic disruptions. Genome-wide coding and noncoding (nc) RNA expression in liver and muscle from 21-month-old prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30-90 of gestation; term: 147 days) and control females were compared. Prenatal T (1) induced differential expression of messenger RNAs (mRNAs) in liver (15 down, 17 up) and muscle (66 down, 176 up) (false discovery rate < 0.05, absolute log2 fold change > 0.5); (2) downregulated mitochondrial pathway genes in liver and muscle; (3) downregulated hepatic lipid catabolism and peroxisome proliferator-activated receptor (PPAR) signaling gene pathways; (4) modulated noncoding RNA (ncRNA) metabolic processes gene pathway in muscle; and (5) downregulated 5 uncharacterized long noncoding RNA (lncRNA) in the muscle but no ncRNA changes in the liver. Correlation analysis showed downregulation of lncRNAs LOC114112974 and LOC105607806 was associated with decreased TPK1, and LOC114113790 with increased ZNF470 expression. Orthogonal projections to latent structures discriminant analysis identified mRNAs HADHA and SLC25A45, and microRNAs MIR154A, MIR25, and MIR487B in the liver and ARIH1 and ITCH and miRNAs MIR369, MIR10A, and MIR10B in muscle as potential biomarkers of prenatal T excess. These findings suggest downregulation of mitochondria, lipid catabolism, and PPAR signaling genes in the liver and dysregulation of mitochondrial and ncRNA gene pathways in muscle are contributors of lipotoxic and insulin-resistant hepatic and muscle phenotype. Gestational T excess programming of metabolic dysfunctions involve tissue-specific ncRNA-modulated transcriptional changes.
Collapse
Affiliation(s)
- Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Muraly Puttabyatappa
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | | | - John Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Joseph N Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Robert C Thompson
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
- Correspondence: Vasantha Padmanabhan, PhD, MS, Department of Pediatrics, University of Michigan, 7510 MSRB1, 1150 W Medical Center Dr, Ann Arbor, MI 48019-5718, USA.
| |
Collapse
|
11
|
Zhang J, Zhang J, Cong S, Feng J, Pan L, Zhu Y, Zhang A, Ma J. Transcriptome profiling of lncRNA and co-expression network in the vaginal epithelial tissue of women with lubrication disorders. PeerJ 2021; 9:e12485. [PMID: 34824921 PMCID: PMC8590395 DOI: 10.7717/peerj.12485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 10/22/2021] [Indexed: 12/14/2022] Open
Abstract
Background Vaginal lubrication is a crucial physiological response that occurs at the beginning of sexual arousal. However, research on lubrication disorders (LD) is still in its infancy, and the role of long non-coding RNAs (lncRNAs) in LD remains unclear. This study aimed to explore the function of lncRNAs in the pathogenesis of vaginal LD. Methods The expression profiles of LD and normal control (NC) lncRNAs were examined using next-generation sequencing (NGS), and eight selected differentially expressed lncRNAs were verified by quantitative real-time PCR. We conducted GO annotation and KEGG pathway enrichment analyses to determine the principal functions of significantly deregulated genes. LncRNA-mRNA co-expression and protein-protein interaction (PPI) networks were constructed and the lncRNA transcription factors (TFs) were predicted. Results From the results, we identified 181,631 lncRNAs and 145,224 mRNAs in vaginal epithelial tissue. Subsequently, our preliminary judgment revealed a total of 499 up-regulated and 337 down-regulated lncRNAs in LD. The top three enriched GO items of the dysregulated lncRNAs included the following significant terms: “contractile fiber part,” “actin filament-based process,” and “contractile fiber”. The most enriched pathways were “cell-extracellular matrix interactions,” “muscle contraction,” “cell-cell communication,” and “cGMP-PKG signaling pathway”. Our results also showed that the lncRNA-mRNA co-expression network was a powerful platform for predicting lncRNA functions. We determined the three hub genes, ADCY5, CXCL12, and NMU, using PPI network construction and analysis. A total of 231 TFs were predicted with RHOXF1, SNAI2, ZNF354C and TBX15 were suspected to be involved in the mechanism of LD. Conclusion In this study, we constructed the lncRNA-mRNA co-expression network, predicted the lncRNA TFs, and comprehensively analyzed lncRNA expression profiles in LD, providing a basis for future studies on LD clinical biomarkers and therapeutic targets. Further research is also needed to fully determine lncRNA’s role in LD development.
Collapse
Affiliation(s)
- Jingjing Zhang
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Jing Zhang
- Jiangsu Health Vocational College, Nanjing, China
| | - Shengnan Cong
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Jingyi Feng
- High School Affiliated to Nanjing Normal University International Department, Nanjing, China
| | - Lianjun Pan
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yuan Zhu
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Aixia Zhang
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Jiehua Ma
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| |
Collapse
|
12
|
李 玉, 陈 婕, 罗 倩, 谈 勇. Research progress in lncRNA and its action as ceRNA in ovarian function as well as the relevant diseases. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:745-752. [PMID: 34382592 PMCID: PMC10930132 DOI: 10.11817/j.issn.1672-7347.2021.200622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 11/03/2022]
Abstract
Long chain non-encoding RNA (lncRNA) can affect gene expression through transcription, post transcriptional regulation and epigenetic modification, and it is involved in regulating ovarian physiological function. LncRNA, as a competitive endogenous RNA, can affect the expression of target gene mRNA by competitively binding microRNA (miRNA), which are called lncRNA/miRNA/mRNA regulatory network. It plays an important role in the regulation of ovarian physiological function and the occurrence and development of ovarian reproductive disorders, expecting to become a new target and diagnostic index for the treatment of reproductive disorders.
Collapse
Affiliation(s)
| | | | | | - 勇 谈
- 谈勇,, ORCID: 0000-0003-3629-1789
| |
Collapse
|
13
|
Zhao Y, Ma S, Cui Z, Li S, Chen Y, Yin Y, Yin Z. The relationship between LncRNAs and lung adenocarcinoma as well as their ceRNA network. Cancer Biomark 2021; 31:165-176. [PMID: 33896828 DOI: 10.3233/cbm-203078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND More and more studies have shown that long non-coding RNA (LncRNA) as a competing endogenous RNA (ceRNA) plays an important role in lung cancer. Therefore, we analyzed the RNA expression profiles of 82 lung cancer patients which were all from Gene Expression Omnibus (GEO). METHODS Firstly, we used BLASTN (evalue = 1e-10) to annotate the gene sets, performed in-group correction and batched normalization of the three data sets with R. Secondly, we used the limma and sva packages to compare tumor tissues with normal tissues. Then through WGCNA, we obtained the 4 gene modules most related to the trait. RESULTS We intersected the genes of above 4 modules with the differential expression genes: 28 LncRNAs (up: 5, down: 23) and 265 mRNAs (up:11, down: 254). Based on these genes, we picked up 6 LncRNAs (CCDC39, FAM182A, SRGAP3-AS2, ADAMTS9-AS2, AC020907.2, SFTA1P), then set and visualized the LncRNA-miRNA-mRNA ceRNA network with 12 miRNAs related to 12 mRNAs. Finally, we performed downstream analysis of 265 mRNAs by Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and Protein-Protein Interaction (PPI) network. CONCLUSION After analyzing, we think this study provides a new direction for basic and clinical research related to LAD, and is expected to provide new targets for early diagnosis, prognostic evaluation and clinical treatment of lung cancer.
Collapse
Affiliation(s)
- Yuxin Zhao
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China.,China Medical University, Shenyang North New Area, Shenyang, Liaoning, China
| | - Shuwen Ma
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China.,China Medical University, Shenyang North New Area, Shenyang, Liaoning, China
| | - Zhigang Cui
- School of Nursing, China Medical University, Shenyang, Liaoning, China
| | - Sixuan Li
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Yao Chen
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China
| | - Yu Yin
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China
| | - Zhihua Yin
- China Medical University, Shenyang North New Area, Shenyang, Liaoning, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China.,Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang North New Area, Shenyang, Liaoning, China
| |
Collapse
|
14
|
López-Camarillo C, Ruíz-García E, Salinas-Vera YM, Silva-Cázares MB, Hernández-de la Cruz ON, Marchat LA, Gallardo-Rincón D. Deciphering the Long Non-Coding RNAs and MicroRNAs Coregulation Networks in Ovarian Cancer Development: An Overview. Cells 2021; 10:1407. [PMID: 34204094 PMCID: PMC8227049 DOI: 10.3390/cells10061407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023] Open
Abstract
Non-coding RNAs are emergent elements from the genome, which do not encode for proteins but have relevant cellular functions impacting almost all the physiological processes occurring in eukaryotic cells. In particular, microRNAs and long non-coding RNAs (lncRNAs) are a new class of small RNAs transcribed from the genome, which modulate the expression of specific genes at transcriptional and posttranscriptional levels, thus adding a new regulatory layer in the flux of genetic information. In cancer cells, the miRNAs and lncRNAs interactions with its target genes and functional pathways are deregulated as a consequence of epigenetic and genetic alterations occurring during tumorigenesis. In this review, we summarize the actual knowledge on the interplay of lncRNAs with its cognate miRNAs and mRNAs pairs, which interact in coregulatory networks with a particular emphasis on the mechanisms underlying its oncogenic behavior in ovarian cancer. Specifically, we reviewed here the evidences unraveling the relevant roles of lncRNAs/miRNAs pairs in altered regulation of cell migration, angiogenesis, therapy resistance, and Warburg effect. Finally, we also discussed its potential clinical implications in ovarian cancer and related endocrine disease therapies.
Collapse
Affiliation(s)
- César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, 03100 CDMX, Mexico;
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
| | - Erika Ruíz-García
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, 14080 CDMX, Mexico
| | | | - Macrina B. Silva-Cázares
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, 78700 San Luis Potosí, Mexico;
| | | | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, 07340 CDMX, Mexico;
| | - Dolores Gallardo-Rincón
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, 14080 CDMX, Mexico
| |
Collapse
|
15
|
Zhou M, Liu X, Qiukai E, Shang Y, Zhang X, Liu S, Zhang X. Long non-coding RNA Xist regulates oocyte loss via suppressing miR-23b-3p/miR-29a-3p maturation and upregulating STX17 in perinatal mouse ovaries. Cell Death Dis 2021; 12:540. [PMID: 34035229 PMCID: PMC8149765 DOI: 10.1038/s41419-021-03831-4] [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: 01/06/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023]
Abstract
The fecundity of female mammals is resolved by the limited size of the primordial follicle (PF) pool formed perinatally. The establishment of PF pool is accompanied by a significant programmed oocyte death. Long non-coding RNAs (lncRNA) are central modulators in regulating cell apoptosis or autophagy in multiple diseases, however, the significance of lncRNAs governing perinatal oocyte loss remains unknown. Here we find that Yin-Yang 1 (YY1) directly binds to the lncRNA X-inactive-specific transcript (Xist) promoter and facilitates Xist expression in the perinatal mouse ovaries. Xist is highly expressed in fetal ovaries and sharply downregulated along with the establishment of PF pool after birth. Gain or loss of function analysis reveals that Xist accelerates oocyte autophagy, mainly through binding to pre-miR-23b or pre-miR-29a in the nucleus and preventing the export of pre-miR-23b/pre-miR-29a to the cytoplasm, thus resulting in decreased mature of miR-23b-3p/miR-29a-3p expression and upregulation miR-23b-3p/miR-29a-3p co-target, STX17, which is essential for timely control of the degree of oocyte death in prenatal mouse ovaries. Overall, these findings identify Xist as a key non-protein factor that can control the biogenesis of miR-23b-3p/miR-29a-3p, and this YY1-Xist-miR-23b-3p/miR-29a-3p-STX17 regulatory axis is responsible for perinatal oocyte loss through autophagy.
Collapse
Affiliation(s)
- Meng Zhou
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Xiaoqiu Liu
- grid.89957.3a0000 0000 9255 8984Department of Microbiology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166 Nanjing, China
| | - E. Qiukai
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Yanxing Shang
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Xiaoqian Zhang
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Shuting Liu
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Xuesen Zhang
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| |
Collapse
|
16
|
Long noncoding RNAs as a piece of polycystic ovary syndrome puzzle. Mol Biol Rep 2021; 48:3845-3851. [PMID: 33993404 DOI: 10.1007/s11033-021-06196-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/28/2021] [Indexed: 10/21/2022]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder and affects 5-10% of reproductive-age women. Chronic anovulation, polycystic ovaries, and hyperandrogenism are the important features of this syndrome. Furthermore, hyperinsulinemia and central obesity are frequent in PCOS women. In recent years, noncoding RNAs detection provided new ideas to explain the etiology of female reproductive disorders. Long noncoding RNAs (lncRNAs) as a subset of noncoding RNAs are associated with the pathogenesis of manifold reproductive-related disorders. Various investigations emphasized the potential involvement of lncRNAs in PCOS development. Therefore, in this paper, we will summarize the function of numerous lncRNAs in the apoptosis and proliferation of granulosa cells (GCs), insulin resistance (IR), and steroidogenesis in PCOS.
Collapse
|
17
|
Sebastian-Leon P, Devesa-Peiro A, Aleman A, Parraga-Leo A, Arnau V, Pellicer A, Diaz-Gimeno P. Transcriptional changes through menstrual cycle reveal a global transcriptional derepression underlying the molecular mechanism involved in the window of implantation. Mol Hum Reprod 2021; 27:6217366. [PMID: 33830236 DOI: 10.1093/molehr/gaab027] [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: 09/24/2020] [Revised: 03/27/2021] [Indexed: 12/20/2022] Open
Abstract
The human endometrium is a dynamic tissue that only is receptive to host the embryo during a brief time in the middle secretory phase, called the window of implantation (WOI). Despite its importance, regulation of the menstrual cycle remains incompletely understood. The aim of this study was to characterize the gene cooperation and regulation of menstrual cycle progression, to dissect the molecular complexity underlying acquisition of endometrial receptivity for a successful pregnancy, and to provide the scientific community with detailed gene co-expression information throughout the menstrual cycle on a user-friendly web-tool database. A retrospective gene co-expression analysis was performed based on the endometrial receptivity array (ERarray) gene signature from 523 human endometrial samples collected across the menstrual cycle, including during the WOI. Gene co-expression analysis revealed the WOI as having the significantly smallest proportion of negative correlations for transcriptional profiles associated with successful pregnancies compared to other cycle stages, pointing to a global transcriptional derepression being involved in acquisition of endometrial receptivity. Regulation was greatest during the transition between proliferative and secretory endometrial phases. Further, we prioritized nuclear hormone receptors as major regulators of this derepression and proved that some genes and transcription factors involved in this process were dysregulated in patients with recurrent implantation failure. We also compiled the wealth of gene co-expression data to stimulate hypothesis-driven single-molecule endometrial studies in a user-friendly database: Menstrual Cycle Gene Co-expression Network (www.menstrualcyclegcn.com). This study revealed a global transcriptional repression across the menstrual cycle, which relaxes when the WOI opens for transcriptional profiles associated with successful pregnancies. These findings suggest that a global transcriptional derepression is needed for embryo implantation and early development.
Collapse
Affiliation(s)
- P Sebastian-Leon
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - A Devesa-Peiro
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Department of Pediatrics, Obstetrics and Gynaecology, University of Valencia, Valencia, Spain
| | - A Aleman
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - A Parraga-Leo
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Department of Pediatrics, Obstetrics and Gynaecology, University of Valencia, Valencia, Spain
| | - V Arnau
- Bioinformatics, Escuela Técnica Superior de Ingeniería, Universidad de Valencia, Burjassot, Spain.,Institute for Integrative Systems Biology (I2SysBio), Universidad de Valencia-Consejo Superior de Investigaciones Científicas (CSIC), C/Catedrático Agustín Escardino Benlloch, Paterna, Spain
| | - A Pellicer
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Department of Pediatrics, Obstetrics and Gynaecology, University of Valencia, Valencia, Spain.,Reproductive Medicine, IVI-RMA IVI Rome, Rome, Italy
| | - P Diaz-Gimeno
- Department of Genomic & Systems Reproductive Medicine, IVI-RMA IVI Foundation-Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
18
|
Ma Y, Ma L, Cao Y, Zhai J. Construction of a ceRNA-based lncRNA-mRNA network to identify functional lncRNAs in polycystic ovarian syndrome. Aging (Albany NY) 2021; 13:8481-8496. [PMID: 33714202 PMCID: PMC8034915 DOI: 10.18632/aging.202659] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder in women of childbearing age. Recent studies suggest important roles for lncRNAs in PCOS development. Based on the hypothesis that lncRNAs are able to regulate mRNA functions by competitive binding to shared miRNAs, the present work sought to construct a PCOS-related lncRNA-mRNA network (PCLMN) to identify key lncRNAs with dysregulated expression and potential prognostic and therapeutic relevance. A global background network was constructed after retrieving lncRNA-miRNA and miRNA-mRNA pairs from the lncRNASNP2, miRTarBase and StarBase databases. Based on gene expression profiles from ovarian granulosa cells from PCOS patients and controls in the GEO’s GSE95728 dataset, the PCLMN was then constructed by applying hypergeometric testing. Using topological analysis, we identified 3 lncRNAs (LINC00667, AC073172.1 and H19) ranking within the top-ten gene lists for all three centrality measures. We then explored their subcellular localization, performed functional module analyses, and identified 4 sex hormone-related transcription factors as potential regulators of their expression. Significant associations with inflammation, oxidative stress, and apoptosis-related processes and pathways were revealed for the key lncRNAs in our PCMLN. Further studies verifying the mRNA/lncRNA relationships identified herein are needed to clarify their clinical significance.
Collapse
Affiliation(s)
- Yue Ma
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linna Ma
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yurong Cao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Zhai
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Tu M, Wu Y, Mu L, Zhang D. Long non-coding RNAs: novel players in the pathogenesis of polycystic ovary syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:173. [PMID: 33569475 PMCID: PMC7867878 DOI: 10.21037/atm-20-5044] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a class of transcripts (>200 nucleotides) lacking protein-coding capacity. Based on the complex three-dimensional structure, lncRNAs are involved in many biological processes and can regulate the expression of target genes at chromatin modification, transcriptional and post-transcriptional levels. LncRNAs have been studied in multiple diseases but little is known about their role(s) in polycystic ovary syndrome (PCOS), the most common endocrinological disorder in reproductive-aged women around the world. In this review, we characterized and explored the potential mechanisms of lncRNAs in the pathogenesis of PCOS. We found that lncRNAs play a molecular role in PCOS mainly by functioning as the competitive endogenous RNA (ceRNA) and are significantly correlated with some clinical phenotypes. We summarized in detail regarding aberrant lncRNAs in different specimens of women with PCOS [i.e., granulosa cells (GCs), cumulus cells (CCs), follicular fluid (FF), peripheral blood] and various PCOS rodent models [i.e., dehydroepiandrosterone (DHEA) and letrozole induced models]. In clinical practice, detection of lncRNAs in serum might enable early diagnosis. Furthermore, new lncRNA-based classifications might be emerging as potent predictors of a particular phenotype in PCOS. Overall, we proposed new insights for the application of precision medicine approaches to the management of PCOS.
Collapse
Affiliation(s)
- Mixue Tu
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Yiqing Wu
- Women's Reproductive Health Research Key Laboratory of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liangshan Mu
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Dan Zhang
- Women's Reproductive Health Research Key Laboratory of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Huang J, Zhao J, Geng X, Chu W, Li S, Chen ZJ, Du Y. Long non-coding RNA lnc-CCNL1-3:1 promotes granulosa cell apoptosis and suppresses glucose uptake in women with polycystic ovary syndrome. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:614-628. [PMID: 33552682 PMCID: PMC7819816 DOI: 10.1016/j.omtn.2020.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disease in premenopausal women. Long non-coding RNAs (lncRNAs) constitute important factors in numerous biological processes. However, their roles in PCOS pathogenesis require further clarification. Our study aims to elucidate the roles of lncRNA lnc-CCNL1-3:1 (CCNL) in PCOS. CCNL expression in human luteinized granulosa cells (hLGCs) derived from women with and without PCOS was detected. The full length of CCNL was obtained by 5' and 3' rapid amplification of cDNA ends. CCNL roles in granulosa cell apoptosis, mitochondrial function, and glucose uptake were evaluated. The binding relationship between CCNL and forkhead box O1 (FOXO1) was determined by RPISeq, RNA immunoprecipitation, subcellular fractionation, and immunofluorescence. In KGN cells and hLGCs, CCNL overexpression upregulated FOXO1 expression, promoted cell apoptosis, reduced glucose transport capability, and impaired mitochondrial function, and these effects were partially abolished by silencing FOXO1. The interaction of CCNL with FOXO1 might prevents FOXO1 exclusion from the nucleus and subsequent degradation in the cytosol. We determined that CCNL serve as a facilitator in the processes of PCOS. CCNL might participate in PCOS pathologies such as follicular atresia and insulin resistance.
Collapse
Affiliation(s)
- Jiayu Huang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jun Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Xueying Geng
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Weiwei Chu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.,Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong Provincial Key Laboratory of Reproductive Medicine, No. 157 Jingliu Road, Jinan 250001, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| |
Collapse
|
21
|
Hong G, Wu H, Ma ST, Su Z. Catechins from oolong tea improve uterine defects by inhibiting STAT3 signaling in polycystic ovary syndrome mice. Chin Med 2020; 15:125. [PMID: 33292347 PMCID: PMC7708239 DOI: 10.1186/s13020-020-00405-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background It is showed that inflammation is causative factor for PCOS, leading to a decline in ovarian fertility. Previous studies have reported that tea consumption can reduce the incidence of ovarian cancer. We speculate that catechins from oolong tea (Camellia sinensis (L.) O. Kuntze) may have a potential therapeutic effect on PCOS. This study aims to investigate the effects of oolong tea catechins on the uterus of polycystic ovary syndrome (PCOS) mice induced by insulin combined with human chorionic gonadotropin (hCG). Methods Sixty female mice were divided into 6 groups (n = 10): model, model + Metformin 200 mg/kg, model + catechins 25 mg/kg, model + catechins 50 mg/kg, and model + catechins 100 mg/kg. Another forty female mice were divided into 4 groups (n = 10): control, control + catechins 100 mg/kg, model, and model + catechins 100 mg/kg. Ovarian and uterine weight coefficients, sex hormone levels, glucose metabolism and insulin resistance, and ovarian and uterine pathology were examined. Changes in NF-κB-mediated inflammation, MMP2 and MMP9 expressions, and STAT3 signaling were evaluated in the uterus of mice. Results Catechins could effectively reduce the ovarian and uterine organ coefficients, reduce the levels of E2, FSH and LH in the blood and the ratio of LH/FSH, and improve glucose metabolism and insulin resistance in PCOS mice induced by insulin combined with hCG. In addition, catechins could significantly down-regulated the expression of p-NF-κB p65 in the uterus and the protein expressions of the pro-inflammatory factors (IL-1β, IL-6, and TNF-α). The expressions of mmp2 and mmp9 associated with matrix degradation in uterine tissue were also significantly down-regulated by catechins. Further, catechins significantly reduced the expression of p-STAT3 and increased the expression of p-IRS1 and p-PI3K in the uterus of PCOS mice. Conclusion Catechins from oolong tea can alleviate ovarian dysfunction and insulin resistance in PCOS mice by inhibiting uterine inflammation and matrix degradation via inhibiting p-STAT3 signaling.
Collapse
Affiliation(s)
- Ge Hong
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Biomedical Material, Tianjin, 300192, China.,Life and Health College, Anhui Science and Technology University, Fengyang, 233100, China
| | - Hao Wu
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 200192, China
| | - Shi-Tang Ma
- Life and Health College, Anhui Science and Technology University, Fengyang, 233100, China.
| | - Zhe Su
- Tianjin Institute for Drug Control, Tianjin, 300000, China
| |
Collapse
|
22
|
Geng X, Zhao J, Huang J, Li S, Chu W, Wang WS, Chen ZJ, Du Y. lnc-MAP3K13-7:1 Inhibits Ovarian GC Proliferation in PCOS via DNMT1 Downregulation-Mediated CDKN1A Promoter Hypomethylation. Mol Ther 2020; 29:1279-1293. [PMID: 33212300 PMCID: PMC7934583 DOI: 10.1016/j.ymthe.2020.11.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine-related disease and global cause of infertility that is associated with abnormal folliculogenesis. Inhibited granulosa cell (GC) proliferation is recognized as a key factor that underlies aberrant follicle maturation. Many epigenetic landscape modifications have been characterized in PCOS patients. However, the epigenetic regulation pathways in follicular dysplasia are not completely understood. In this study, we reported a novel mechanism of DNA hypomethylation induced by long non-coding RNAs (lncRNAs) and its function in cell cycle progression. We observed that lnc-MAP3K13-7:1 was highly expressed in GCs from patients with PCOS, with concomitant global DNA hypomethylation, decreased DNA methyltransferase 1 (DNMT1) expression, and increased cyclin-dependent kinase inhibitor 1A (CDKN1A, p21) expression. In KGN cells, lnc-MAP3K13-7:1 overexpression resulted in cell cycle arrest in the G0/G1 phase, as well as the molecular inhibition and genetic silencing of DNMT1. Mechanistically, lnc-MAP3K13-7:1 inhibited DNMT1 expression by acting as a protein-binding scaffold and inducing ubiquitin-mediated DNMT1 protein degradation. Moreover, DNMT1-dependent CDKN1A promoter hypomethylation increased CDKN1A transcription, resulting in attenuated GC growth. Our work uncovered a novel and essential mechanism through which lnc-MAP3K13-7:1-dependent DNMT1 inhibition regulates CDKN1A/p21 expression and inhibits GC proliferation.
Collapse
Affiliation(s)
- Xueying Geng
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jun Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jiayu Huang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Weiwei Chu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China; Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
23
|
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
|
24
|
Tu J, Chen Y, Li Z, Yang H, Chen H, Yu Z. Long non-coding RNAs in ovarian granulosa cells. J Ovarian Res 2020; 13:63. [PMID: 32503679 PMCID: PMC7275442 DOI: 10.1186/s13048-020-00663-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Granulosa cells (GCs) are somatic cells surrounding oocytes within follicles and are essential for folliculogenesis. Pathological changes in GCs are found in several ovarian disorders. Recent reports have indicated that long non-coding RNAs (lncRNAs), which modulate gene expression via multiple mechanisms, are key regulators of the normal development of GCs, follicles, and ovaries. In addition, accumulating evidence has suggested that lncRNAs can be utilized as biomarkers for the diagnosis and prognosis of GC-related diseases, such as polycystic ovary syndrome (PCOS) and premature ovarian insufficiency (POI). Therefore, lncRNAs not only play a role in GCs that are involved in normal folliculogenesis, but they may also be considered as potential candidate biomarkers and therapeutic targets in GCs under pathological conditions. In the future, a detailed investigation of the in vivo delivery or targeting of lncRNAs and large-cohort-validation of the clinical applicability of lncRNAs is required.
Collapse
Affiliation(s)
- Jiajie Tu
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China. .,Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui province, China.
| | - Yu Chen
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China
| | - Zhe Li
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China
| | - Huan Yang
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China
| | - He Chen
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China
| | - Zhiying Yu
- Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 Sungang West Road, Futian District, Shenzhen, 518000, Guangdong province, China.
| |
Collapse
|
25
|
Wu G, Yang Z, Chen Y, Li X, Yang J, Yin T. Downregulation of Lnc-OC1 attenuates the pathogenesis of polycystic ovary syndrome. Mol Cell Endocrinol 2020; 506:110760. [PMID: 32070768 DOI: 10.1016/j.mce.2020.110760] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023]
Abstract
Long non-coding RNAs (lncRNAs) play a vital role in the progression of many human diseases. The aim of this study is to explore the relationship between lncRNA-ovarian cancer associated 1 (Lnc-OC1) and PCOS. In this study, we found that Lnc-OC1 was significantly higher in PCOS granulosa cells (GCs) compared to non-PCOS GCs. Lnc-OC1 knockdown inhibited cell viability and promoted cell apoptosis, expression of aromatase mRNA and production of estradiol in KGN cells. In PCOS mice, Lnc-OC1 promoted the serum insulin release, production of angiogenesis-related factors and IκBα phosphorylation, which could be partially restored by Lnc-OC1 shRNA. These results suggest that Lnc-OC1 plays an important part in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Gengxiang Wu
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Zhe Yang
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Yajie Chen
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Xiaoling Li
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| | - Tailang Yin
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|