1
|
Kobayashi R, Hatada I. Understanding epigenetic regulation in the endometrium - lessons from mouse models with implantation defects. Epigenomics 2025:1-14. [PMID: 40228031 DOI: 10.1080/17501911.2025.2491298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Endometrial function, crucial for successful embryo implantation, is significantly influenced by epigenetic regulation. This review investigates the crucial roles of DNA methylation, histone modifications, chromatin remodeling, and RNA methylation in endometrial receptivity and implantation, based on a survey of recent literature on knockout mouse models with implantation defects. These models illuminate how epigenetic disruptions contribute to implantation failure, a significant human reproductive health concern. DNA methylation and histone modifications modulate endometrial receptivity by affecting gene silencing and chromatin structure, respectively. Chromatin remodeling factors also play a critical role in endometrial dynamics, influencing gene expression. Furthermore, RNA methylation emerges as critical in implantation through transcriptional and translational control. While human studies provide limited epigenetic snapshots, mouse models with suppressed epigenetic regulators reveal direct causal links between epigenetic alterations and implantation failure. Understanding these epigenetic interactions offers potential for novel therapies addressing reproductive disorders.
Collapse
Affiliation(s)
- Ryosuke Kobayashi
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Gunma, Japan
| |
Collapse
|
2
|
Jia S, Zhao F. Decoding Müllerian Duct Epithelial Regionalization. Mol Reprod Dev 2025; 92:e70018. [PMID: 39994938 PMCID: PMC11850963 DOI: 10.1002/mrd.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
Müllerian ducts (MD), also known as paramesonephric ducts, are the primordial anlage of the female reproductive tract organs including the oviduct, uterus, cervix and upper vagina along the craniocaudal axis. Although the general architecture of MD-derived organs is conserved, each organ possesses their unique epithelial structures and cell types to confer their region-specific functions, which collectively coordinate successful fertilization and pregnancy. MD epithelial fate decisions and differentiation along the craniocaudal axis is dependent on spatiotemporal regulation of intrinsic transcription factors and extrinsic signals derived from the mesenchyme. Findings from genetic mouse models, single-cell sequencing studies, and organoid cultures have significantly advanced our understanding of the cellular and molecular mechanisms of MD regionalization. In this review, we first discuss the diversity of epithelial morphologies and cell types in the female reproductive tract organs. Then, we discuss the roles of key transcription factors (Hox, transcriptional cascade driving multiciliogenesis, Foxa2, and P63), signaling pathways (estrogen/ESR1, Wnt/β-catenin, hedgehog, and retinoic acid), and epigenetic factors (microRNAs, chromatin remodeling factors, and histone modification enzymes) in region-specific MD differentiation. Further deciphering molecular mechanisms of MD craniocaudal patterning will open new avenues to improve our strategies for prevention, diagnosis, and treatment of Müllerian anomalies and female reproductive tract disorders.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
3
|
Akaeda S, Aikawa S, Hirota Y. Spatial and molecular anatomy of the endometrium during embryo implantation: a current overview of key regulators of blastocyst invasion. FEBS J 2024; 291:4206-4221. [PMID: 38348632 DOI: 10.1111/febs.17077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/09/2024] [Accepted: 01/23/2024] [Indexed: 10/04/2024]
Abstract
Embryo implantation is composed of three steps: blastocyst apposition, adhesion/attachment and invasion. Blastocyst invasion has been studied less extensively than the other two events. Historically, studies conducted using electron microscopy have shown the removal of epithelial cells in the vicinity of the attached blastocysts in rodents, although the underlying mechanisms have remained unclear. Here, we describe recent studies using mice with uterine-specific gene deletion that demonstrated important roles for nuclear proteins such as progesterone receptor, hypoxia inducible factor and retinoblastoma in the regulation of embryo invasion. In these mouse models, the detachment of the endometrial luminal epithelium, decidualization in the stroma, and the activation of trophoblasts have been found to be important in ensuring embryo invasion. This review summarizes the molecular signaling associated with these cellular events, mainly evidenced by mouse models.
Collapse
Affiliation(s)
- Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Shizu Aikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
4
|
Kobayashi R, Tajika Y, Kohmaru J, Morita S, Horii T, Mizukami Y, Aikawa S, Hirota Y, Hatada I. The histone methyltransferase KMT2D is essential for embryo implantation via regulating precise differentiation of endometrial cells. Cell Death Discov 2024; 10:357. [PMID: 39117610 PMCID: PMC11310208 DOI: 10.1038/s41420-024-02134-9] [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: 05/15/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
Embryo implantation failures are a major challenge in reproductive medicine, but the underlying mechanism remains poorly understood. Successful implantation requires dynamic remodeling of the endometrium through integrated proliferation and differentiation of endometrial cells including luminal epithelial, glandular epithelial, and stromal cells. Conversely, their disruption causes infertility. Spatiotemporal control of transcription is required for these processes; however, the underlying epigenetic regulation is largely unknown. In this study, we examined expression data from the human endometrium during implantation and discovered that expression of the histone lysine methyltransferase KMT2D was significantly suppressed in patients with recurrent implantation failure. Further study revealed that uterine deletion of Kmt2d in mice caused infertility due to implantation failure. Morphological analysis discovered a reduction in the number of uterine glands and aberrant differentiation of the luminal and glandular epithelium into stratified phenotypes in Kmt2d knockout uteri. Administration of leukemia inhibitory factor protein, which is expressed in uterine glands and is essential for implantation, did not rescue implantation failure in Kmt2d knockout mice, suggesting that infertility was not solely due to uterine gland dysfunction. RNA sequencing analysis revealed that Kmt2d knockout uteri displayed suppressed expression of genes involved in ion homeostasis, which may affect the uterine luminal morphology. Our study suggests that KMT2D plays an essential role in facilitating successful embryo implantation by regulating the coordinated differentiation of endometrial cells, providing valuable insights into unexplained implantation failures in women.
Collapse
Affiliation(s)
- Ryosuke Kobayashi
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yuki Tajika
- Department of Anatomy, Gunma University Graduate School of Medicine, Maebashi, Japan
- Gunma Prefectural College of Health Sciences, Maebashi, Japan
| | - Junki Kohmaru
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Sumiyo Morita
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Takuro Horii
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yoichi Mizukami
- Institute of Gene Research, Science Research Center, Yamaguchi University, Yamaguchi, Japan
| | - Shizu Aikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan.
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Gunma, Japan.
| |
Collapse
|
5
|
Siddique R, Gupta G, Mgm J, Kumar A, Kaur H, Ariffin IA, Pramanik A, Almalki WH, Ali H, Shahwan M, Patel N, Murari K, Mishra R, Thapa R, Bhat AA. Targeting notch-related lncRNAs in cancer: Insights into molecular regulation and therapeutic potential. Pathol Res Pract 2024; 257:155282. [PMID: 38608371 DOI: 10.1016/j.prp.2024.155282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
Cancer is a group of diseases marked by unchecked cell proliferation and the ability for the disease to metastasize to different body areas. Enhancements in treatment and early detection are crucial for improved outcomes. LncRNAs are RNA molecules that encode proteins and have a length of more than 200 nucleotides. LncRNAs are crucial for chromatin architecture, gene regulation, and other cellular activities that impact both normal growth & pathological processes, even though they are unable to code for proteins. LncRNAs have emerged as significant regulators in the study of cancer biology, with a focus on their intricate function in the Notch signaling pathway. The imbalance of this pathway is often linked to a variety of malignancies. Notch signaling is essential for cellular functions like proliferation, differentiation, and death. The cellular response is shaped by these lncRNAs through their modulation of essential Notch pathway constituents such as receptors, ligands, and downstream effectors around it. Furthermore, a variety of cancer types exhibit irregular expression of Notch-related lncRNAs, underscoring their potential use as therapeutic targets and diagnostic markers. Gaining an understanding of the molecular processes behind the interaction between the Notch pathway and lncRNAs will help you better understand the intricate regulatory networks that control the development of cancer. This can open up new possibilities for individualized treatment plans and focused therapeutic interventions. The intricate relationships between lncRNAs & the Notch pathway in cancer are examined in this review.
Collapse
Affiliation(s)
- Raihan Siddique
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Johar Mgm
- Management and Science University (MSU), Shah Alam, Selangor 40100 MSU, Malaysia
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand- 831001, India
| | - I A Ariffin
- Management and Science University (MSU), Shah Alam, Selangor 40100 MSU, Malaysia
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Neeraj Patel
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Krishna Murari
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Riya Mishra
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India.
| |
Collapse
|
6
|
Mohebbi H, Esbati R, Hamid RA, Akhavanfar R, Radi UK, Siri G, Yazdani O. EZH2-interacting lncRNAs contribute to gastric tumorigenesis; a review on the mechanisms of action. Mol Biol Rep 2024; 51:334. [PMID: 38393645 DOI: 10.1007/s11033-024-09237-7] [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: 11/13/2023] [Accepted: 01/10/2024] [Indexed: 02/25/2024]
Abstract
Gastric cancer (GC) remains one of the deadliest malignancies worldwide, demanding new targets to improve its diagnosis and treatment. Long non-coding RNAs (lncRNAs) are dysregulated through gastric tumorigenesis and play a significant role in GC progression and development. Recent studies have revealed that lncRNAs can interact with histone-modifying polycomb protein, enhance Zeste Homolog 2 (EZH2), and mediate its site-specific functioning. EZH2, which functions as an oncogene in GC, is the catalytic subunit of the PRC2 complex that induces H3K27 trimethylation and epigenetically represses gene expression. EZH2-interacting lncRNAs can recruit EZH2 to the promoter regions of various tumor suppressor genes and cause their transcriptional deactivation via histone methylation. The interactions between EZH2 and this lncRNA modulate different processes, such as cell cycle, cell proliferation and growth, migration, invasion, metastasis, and drug resistance, in vitro and in vivo GC models. Therefore, EZH2-interacting lncRNAs are exciting targets for developing novel targeted therapies for GC. Subsequently, this review aims to focus on the roles of these interactions in GC progression to understand the therapeutic value of EZH2-interacting lncRNAs further.
Collapse
Affiliation(s)
- Hossein Mohebbi
- Kermanshah University of medical sciences, International branch, Kermanshah, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | | | - Roozbeh Akhavanfar
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Goli Siri
- Department of Internal Medicine, Amir Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
7
|
Fang X, Nie L, Putluri S, Ni N, Bartholin L, Li Q. Sertoli Cell-Specific Activation of Transforming Growth Factor Beta Receptor 1 Leads to Testicular Granulosa Cell Tumor Formation. Cells 2023; 12:2717. [PMID: 38067144 PMCID: PMC10706251 DOI: 10.3390/cells12232717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The transforming growth factor β (TGFβ) superfamily, consisting of protein ligands, receptors, and intracellular SMAD transducers, regulates fundamental biological processes and cancer development. Our previous study has shown that sustained activation of TGFβ receptor 1 (TGFBR1) driven by anti-Mullerian hormone receptor type 2 (Amhr2)-Cre in the mouse testis induces the formation of testicular granulosa cell tumors (TGCTs). As Amhr2-Cre is expressed in both Sertoli cells and Leydig cells, it remains unclear whether the activation of TGFBR1 in Sertoli cells alone is sufficient to induce TGCT formation. Therefore, the objective of this study was to determine whether Sertoli cell-activation of TGFBR1 drives oncogenesis in the testis. Our hypothesis was that overactivation of TGFBR1 in Sertoli cells would promote their transdifferentiation into granulosa-like cells and the formation of TGCTs. To test this hypothesis, we generated mice harboring constitutive activation of TGFBR1 in Sertoli cells using anti-Mullerian hormone (Amh)-Cre. Disorganized seminiferous tubules and tumor nodules were found in TGFBR1CA; Amh-Cre mice. A histological analysis showed that Sertoli cell-specific activation of TGFBR1 led to the development of neoplasms resembling granulosa cell tumors, which derailed spermatogenesis. Moreover, TGCTs expressed granulosa cell markers including FOXL2, FOXO1, and INHA. Using a dual fluorescence reporter line, the membrane-targeted tdTomato (mT)/membrane-targeted EGFP (mG) mouse, we provided evidence that Sertoli cells transdifferentiated toward a granulosa cell fate during tumorigenesis. Thus, our findings indicate that Sertoli cell-specific activation of TGFBR1 leads to the formation of TGCTs, supporting a key contribution of Sertoli cell reprogramming to the development of this testicular malignancy in our model.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Linfeng Nie
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Satwikreddy Putluri
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Laurent Bartholin
- INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Lyon 1, F-69000 Lyon, France
- Centre Léon Bérard, F-69008 Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
8
|
Zhang X, Cao Q, Rajachandran S, Grow EJ, Evans M, Chen H. Dissecting mammalian reproduction with spatial transcriptomics. Hum Reprod Update 2023; 29:794-810. [PMID: 37353907 PMCID: PMC10628492 DOI: 10.1093/humupd/dmad017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/15/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Mammalian reproduction requires the fusion of two specialized cells: an oocyte and a sperm. In addition to producing gametes, the reproductive system also provides the environment for the appropriate development of the embryo. Deciphering the reproductive system requires understanding the functions of each cell type and cell-cell interactions. Recent single-cell omics technologies have provided insights into the gene regulatory network in discrete cellular populations of both the male and female reproductive systems. However, these approaches cannot examine how the cellular states of the gametes or embryos are regulated through their interactions with neighboring somatic cells in the native tissue environment owing to tissue disassociations. Emerging spatial omics technologies address this challenge by preserving the spatial context of the cells to be profiled. These technologies hold the potential to revolutionize our understanding of mammalian reproduction. OBJECTIVE AND RATIONALE We aim to review the state-of-the-art spatial transcriptomics (ST) technologies with a focus on highlighting the novel biological insights that they have helped to reveal about the mammalian reproductive systems in the context of gametogenesis, embryogenesis, and reproductive pathologies. We also aim to discuss the current challenges of applying ST technologies in reproductive research and provide a sneak peek at what the field of spatial omics can offer for the reproduction community in the years to come. SEARCH METHODS The PubMed database was used in the search for peer-reviewed research articles and reviews using combinations of the following terms: 'spatial omics', 'fertility', 'reproduction', 'gametogenesis', 'embryogenesis', 'reproductive cancer', 'spatial transcriptomics', 'spermatogenesis', 'ovary', 'uterus', 'cervix', 'testis', and other keywords related to the subject area. All relevant publications until April 2023 were critically evaluated and discussed. OUTCOMES First, an overview of the ST technologies that have been applied to studying the reproductive systems was provided. The basic design principles and the advantages and limitations of these technologies were discussed and tabulated to serve as a guide for researchers to choose the best-suited technologies for their own research. Second, novel biological insights into mammalian reproduction, especially human reproduction revealed by ST analyses, were comprehensively reviewed. Three major themes were discussed. The first theme focuses on genes with non-random spatial expression patterns with specialized functions in multiple reproductive systems; The second theme centers around functionally interacting cell types which are often found to be spatially clustered in the reproductive tissues; and the thrid theme discusses pathological states in reproductive systems which are often associated with unique cellular microenvironments. Finally, current experimental and computational challenges of applying ST technologies to studying mammalian reproduction were highlighted, and potential solutions to tackle these challenges were provided. Future directions in the development of spatial omics technologies and how they will benefit the field of human reproduction were discussed, including the capture of cellular and tissue dynamics, multi-modal molecular profiling, and spatial characterization of gene perturbations. WIDER IMPLICATIONS Like single-cell technologies, spatial omics technologies hold tremendous potential for providing significant and novel insights into mammalian reproduction. Our review summarizes these novel biological insights that ST technologies have provided while shedding light on what is yet to come. Our review provides reproductive biologists and clinicians with a much-needed update on the state of art of ST technologies. It may also facilitate the adoption of cutting-edge spatial technologies in both basic and clinical reproductive research.
Collapse
Affiliation(s)
- Xin Zhang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiqi Cao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shreya Rajachandran
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward J Grow
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie Evans
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haiqi Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Bi S, Tu Z, Chen D, Zhang S. Histone modifications in embryo implantation and placentation: insights from mouse models. Front Endocrinol (Lausanne) 2023; 14:1229862. [PMID: 37600694 PMCID: PMC10436591 DOI: 10.3389/fendo.2023.1229862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
Embryo implantation and placentation play pivotal roles in pregnancy by facilitating crucial maternal-fetal interactions. These dynamic processes involve significant alterations in gene expression profiles within the endometrium and trophoblast lineages. Epigenetics regulatory mechanisms, such as DNA methylation, histone modification, chromatin remodeling, and microRNA expression, act as regulatory switches to modulate gene activity, and have been implicated in establishing a successful pregnancy. Exploring the alterations in these epigenetic modifications can provide valuable insights for the development of therapeutic strategies targeting complications related to pregnancy. However, our current understanding of these mechanisms during key gestational stages remains incomplete. This review focuses on recent advancements in the study of histone modifications during embryo implantation and placentation, while also highlighting future research directions in this field.
Collapse
Affiliation(s)
- Shilei Bi
- Key Laboratory for Major Obstetric Diseases of Guangdong, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou, China
| | - Zhaowei Tu
- Key Laboratory for Major Obstetric Diseases of Guangdong, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou, China
| | - Dunjin Chen
- Key Laboratory for Major Obstetric Diseases of Guangdong, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou, China
| | - Shuang Zhang
- Key Laboratory for Major Obstetric Diseases of Guangdong, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangzhou, China
| |
Collapse
|
10
|
Sirohi VK, Medrano TI, Kannan A, Bagchi IC, Cooke PS. Uterine-specific Ezh2 deletion enhances stromal cell senescence and impairs placentation, resulting in pregnancy loss. iScience 2023; 26:107028. [PMID: 37360688 PMCID: PMC10285549 DOI: 10.1016/j.isci.2023.107028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Maternal uterine remodeling facilitates embryo implantation, stromal cell decidualization and placentation, and perturbation of these processes may cause pregnancy loss. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that epigenetically represses gene transcription; loss of uterine EZH2 affects endometrial physiology and induces infertility. We utilized a uterine Ezh2 conditional knockout (cKO) mouse to determine EZH2's role in pregnancy progression. Despite normal fertilization and implantation, embryo resorption occurred mid-gestation in Ezh2cKO mice, accompanied by compromised decidualization and placentation. Western blot analysis revealed Ezh2-deficient stromal cells have reduced amounts of the histone methylation mark H3K27me3, causing upregulation of senescence markers p21 and p16 and indicating that enhanced stromal cell senescence likely impairs decidualization. Placentas from Ezh2cKO dams on gestation day (GD) 12 show architectural defects, including mislocalization of spongiotrophoblasts and reduced vascularization. In summary, uterine Ezh2 loss impairs decidualization, increases decidual senescence, and alters trophoblast differentiation, leading to pregnancy loss.
Collapse
Affiliation(s)
- Vijay K. Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Theresa I. Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Athilakshmi Kannan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Indrani C. Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
11
|
Fukui Y, Hirota Y, Aikawa S, Sakashita A, Shimizu-Hirota R, Takeda N, Ishizawa C, Iida R, Kaku T, Hirata T, Hiraoka T, Akaeda S, Matsuo M, Osuga Y. The EZH2-PRC2-H3K27me3 axis governs the endometrial cell cycle and differentiation for blastocyst invasion. Cell Death Dis 2023; 14:320. [PMID: 37198149 DOI: 10.1038/s41419-023-05832-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/19/2023]
Abstract
Infertility occurs in 15% of couples worldwide. Recurrent implantation failure (RIF) is one of the major problems in in vitro fertilization and embryo transfer (IVF-ET) programs, and how to manage patients with RIF to achieve successful pregnancy outcomes remains unresolved. Here, a uterine polycomb repressive complex 2 (PRC2)-regulated gene network was found to control embryo implantation. Our RNA-seq analyses of the human peri-implantation endometrium obtained from patients with RIF and fertile controls revealed that PRC2 components, including its core enzyme enhancer of zeste homolog 2 (EZH2)-catalyzing H3K27 trimethylation (H3K27me3) and their target genes are dysregulated in the RIF group. Although fertility of uterine epithelium-specific knockout mice of Ezh2 (eKO mice) was normal, Ezh2-deleted mice in the uterine epithelium and stroma (uKO mice) exhibited severe subfertility, suggesting that stromal Ezh2 plays a key role in female fertility. The RNA-seq and ChIP-seq analyses revealed that H3K27me3-related dynamic gene silencing is canceled, and the gene expression of cell-cycle regulators is dysregulated in Ezh2-deleted uteri, causing severe epithelial and stromal differentiation defects and failed embryo invasion. Thus, our findings indicate that the EZH2-PRC2-H3K27me3 axis is critical to preparing the endometrium for the blastocyst invasion into the stroma in mice and humans.
Collapse
Affiliation(s)
- Yamato Fukui
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.
| | - Shizu Aikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Akihiko Sakashita
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, 160-0016, Japan
| | - Ryoko Shimizu-Hirota
- Department of Internal Medicine, Center for Preventive Medicine, Keio University School of Medicine, Tokyo, 160-0016, Japan
| | - Norihiko Takeda
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Chihiro Ishizawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Rei Iida
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Tetsuaki Kaku
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Tomoyuki Hirata
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Takehiro Hiraoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Mitsunori Matsuo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| |
Collapse
|
12
|
Role of EZH2 in Uterine Gland Development. Int J Mol Sci 2022; 23:ijms232415665. [PMID: 36555314 PMCID: PMC9779349 DOI: 10.3390/ijms232415665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a core component of polycomb repressive complex 2 that plays a vital role in transcriptional repression of gene expression. Conditional ablation of EZH2 using progesterone receptor (Pgr)-Cre in the mouse uterus has uncovered its roles in regulating uterine epithelial cell growth and stratification, suppressing decidual myofibroblast activation, and maintaining normal female fertility. However, it is unclear whether EZH2 plays a role in the development of uterine glands, which are required for pregnancy success. Herein, we created mice with conditional deletion of Ezh2 using anti-Mullerian hormone receptor type 2 (Amhr2)-Cre recombinase that is expressed in mesenchyme-derived cells of the female reproductive tract. Strikingly, these mice showed marked defects in uterine adenogenesis. Unlike Ezh2 Pgr-Cre conditional knockout mice, deletion of Ezh2 using Amhr2-Cre did not lead to the differentiation of basal-like cells in the uterus. The deficient uterine adenogenesis was accompanied by impaired uterine function and pregnancy loss. Transcriptomic profiling using next generation sequencing revealed dysregulation of genes associated with signaling pathways that play fundamental roles in development and disease. In summary, this study has identified an unrecognized role of EZH2 in uterine gland development, a postnatal event critical for pregnancy success and female fertility.
Collapse
|
13
|
Fang X, Ni N, Wang X, Tian Y, Ivanov I, Rijnkels M, Bayless KJ, Lydon JP, Li Q. EZH2 and Endometrial Cancer Development: Insights from a Mouse Model. Cells 2022; 11:cells11050909. [PMID: 35269532 PMCID: PMC8909840 DOI: 10.3390/cells11050909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 01/26/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive complex 2, plays an important role in cancer development. As both oncogenic and tumor suppressive functions of EZH2 have been documented in the literature, the objective of this study is to determine the impact of Ezh2 deletion on the development and progression of endometrial cancer induced by inactivation of phosphatase and tensin homolog (PTEN), a tumor suppressor gene frequently dysregulated in endometrial cancer patients. To this end, we created mice harboring uterine deletion of both Ezh2 and Pten using Cre recombinase driven by the progesterone receptor (Pgr) promoter. Our results showed reduced tumor burden in Ptend/d; Ezh2d/d mice compared with that of Ptend/d mice during early carcinogenesis. The decreased Ki67 index in EZH2 and PTEN-depleted uteri versus that in PTEN-depleted uteri indicated an oncogenic role of EZH2 during early tumor development. However, mice harboring uterine deletion of both Ezh2 and Pten developed unfavorable disease outcome, accompanied by exacerbated epithelial stratification and heightened inflammatory response. The observed effect was non-cell autonomous and mediated by altered immune response evidenced by massive accumulation of intraluminal neutrophils, a hallmark of endometrial carcinoma in Ptend/d; Ezh2d/d mice during disease progression. Hence, these results reveal dual roles of EZH2 in endometrial cancer development.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (X.F.); (N.N.); (M.R.)
| | - Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (X.F.); (N.N.); (M.R.)
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA;
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (Y.T.); (I.I.)
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (Y.T.); (I.I.)
| | - Monique Rijnkels
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (X.F.); (N.N.); (M.R.)
| | - Kayla J. Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (X.F.); (N.N.); (M.R.)
- Correspondence: ; Tel.: +1-979-862-2009; Fax: +1-979-847-8981
| |
Collapse
|
14
|
Osokine I, Siewiera J, Rideaux D, Ma S, Tsukui T, Erlebacher A. Gene silencing by EZH2 suppresses TGF-β activity within the decidua to avert pregnancy-adverse wound healing at the maternal-fetal interface. Cell Rep 2022; 38:110329. [PMID: 35108527 PMCID: PMC8833843 DOI: 10.1016/j.celrep.2022.110329] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/23/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
A little-appreciated feature of early pregnancy is that embryo implantation and placental outgrowth do not evoke wound-healing responses in the decidua, the specialized endometrial tissue that surrounds the conceptus. Here, we provide evidence that this phenomenon is partly due to an active program of gene silencing mediated by EZH2, a histone methyltransferase that generates repressive histone 3 lysine 27 trimethyl (H3K27me3) histone marks. We find that pregnancies in mice with EZH2-deficient decidual stromal cells frequently fail by mid-gestation, with the decidua showing ectopic myofibroblast formation, peri-embryonic collagen deposition, and gene expression profiles associated with transforming growth factor β (TGF-β)-driven fibroblast activation and fibrogenic extracellular matrix (ECM) remodeling. Analogous responses are observed when the mutant decidua is surgically wounded, while blockade of TGF-β receptor signaling inhibits the defects and improves reproductive outcomes. Together, these results highlight a critical feature of reproductive success and have implications for the context-specific control of TGF-β-mediated wound-healing responses elsewhere in the body.
Collapse
Affiliation(s)
- Ivan Osokine
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue Medical Sciences, S-1057B, San Francisco, CA 94143-0451, USA
| | - Johan Siewiera
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue Medical Sciences, S-1057B, San Francisco, CA 94143-0451, USA
| | - Damon Rideaux
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue Medical Sciences, S-1057B, San Francisco, CA 94143-0451, USA
| | - Stephany Ma
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue Medical Sciences, S-1057B, San Francisco, CA 94143-0451, USA
| | - Tatsuya Tsukui
- Lung Biology Center, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adrian Erlebacher
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue Medical Sciences, S-1057B, San Francisco, CA 94143-0451, USA; Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Program, University of California San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
15
|
Zhang H, Wu J, Li Y, Jin G, Tian Y, Kang S. Identification of Key Differentially Methylated/Expressed Genes and Pathways for Ovarian Endometriosis by Bioinformatics Analysis. Reprod Sci 2021; 29:1630-1643. [PMID: 34671938 DOI: 10.1007/s43032-021-00751-8] [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] [Received: 04/27/2021] [Accepted: 09/25/2021] [Indexed: 12/25/2022]
Abstract
The goal of this study was to identify genes that were differentially methylated and differentially expressed and their related signaling pathways in ovarian endometriosis tissue. First, the DNA methylation and gene expression profiles in the endometrial tissue of patients with ovarian endometriosis were studied using Illumina 450K methylation microarray analysis and the GSE141549 gene expression dataset. Second, differentially methylated and differentially expressed genes, herein referred to as differentially methylated/expressed genes, were identified and protein-protein interaction networks and functional analysis of these genes were determined. Third, qPCR and immunohistochemistry of patient samples was used to confirm the differential expression of a subset of differentially methylated/expressed genes. Finally, the GSE7305 dataset was used confirm the expression profile of differentially methylated/expressed genes and to determine the potential usefulness of these genes for diagnosis of endometriosis. A total of 37 hypermethylated low-expression genes and 66 hypomethylated high-expression genes were identified in ovarian endometriosis patients. Protein-protein interaction and functional analysis highlighted 8 hypermethylated low-expression genes (KRT19, KRT8, ESR1, PRL, SFN, IL20RA, IL2RB, and PAX8) and 4 hypomethylated high-expression genes (CYP11A1, NR5A1, ME1, and GSTM1). Significantly, both of these gene sets had a diagnostic value for patients with ovarian endometriosis. Signaling pathways that were identified included JAK-STAT (involving IL20RA and IL2RB), prolactin (involving PRL and ESR1), Staphylococcus aureus infection (involving KRT19), viral protein interaction with cytokine and cytokine receptor (involving IL20RA and IL2RB), cytokine-cytokine receptor interaction (involving IL20RA and IL2RB), and drug metabolism-cytochrome P450 (involving GSTM1). The differentially methylated/expressed genes and enriched signaling pathways identified in this study are likely to be associated with the process of ovarian endometriosis.
Collapse
Affiliation(s)
- Haibo Zhang
- Department of Gynecology, Hebei Medical University Fourth Hospital, Jiankanglu 12, Shijiazhuang, 050011, People's Republic of China
| | - Jianlei Wu
- Department of Gynecology, Hebei Medical University Fourth Hospital, Jiankanglu 12, Shijiazhuang, 050011, People's Republic of China
| | - Yan Li
- Department of Molecular Biology, Hebei Medical University Fourth Hospital, Shijiazhuang, People's Republic of China
| | - Ge Jin
- Department of Gynecological Oncology, Medical University Fourth Hospital, Shijiazhuang, People's Republic of China
| | - Yunjie Tian
- Department of Gynecology, Hebei Medical University Fourth Hospital, Jiankanglu 12, Shijiazhuang, 050011, People's Republic of China
| | - Shan Kang
- Department of Gynecology, Hebei Medical University Fourth Hospital, Jiankanglu 12, Shijiazhuang, 050011, People's Republic of China.
| |
Collapse
|
16
|
Kim YY, Kim KS, Kim YJ, Kim SW, Kim H, Ku SY. Transcriptome Analyses Identify Potential Key microRNAs and Their Target Genes Contributing to Ovarian Reserve. Int J Mol Sci 2021; 22:10819. [PMID: 34639162 PMCID: PMC8509654 DOI: 10.3390/ijms221910819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Female endocrinological symptoms, such as premature ovarian inefficiency (POI) are caused by diminished ovarian reserve and chemotherapy. The etiology of POI remains unknown, but this can lead to infertility. This has accelerated the search for master regulator genes or other molecules that contribute as enhancers or silencers. The impact of regulatory microRNAs (miRNAs) on POI has gained attention; however, their regulatory function in this condition is not well known. RNA sequencing was performed at four stages, 2-(2 W), 6-(6 W), 15-(15 W), and 20-(20 W) weeks, on ovarian tissue samples and 5058 differentially expressed genes (DEGs) were identified. Gene expression and enrichment were analyzed based on the gene ontology and KEGG databases, and their association with other proteins was assessed using the STRING database. Gene set enrichment analysis was performed to identify the key target genes. The DEGs were most highly enriched in 6 W and 15 W groups. Figla, GDF9, Nobox, and Pou51 were significantly in-creased at 2 W compared with levels at 6 W and 20 W, whereas the expression of Foxo1, Inha, and Taf4b was significantly de-creased at 20 W. Ccnd2 and Igf1 expression was maintained at similar levels in each stage. In total, 27 genes were upregulated and 26 genes interacted with miRNAs; moreover, stage-specific upregulated and downregulated interactions were demonstrated. Increased and decreased miRNAs were identified at each stage in the ovaries. The constitutively expressed genes, Ccnd2 and Igf1, were identified as the major targets of many miRNAs (p < 0.05), and Fshr and Foxo3 interacted with miRNAs, namely mmu-miR-670-3p and mmu-miR-153-3p. miR-26a-5p interacted with Piwil2, and its target genes were downregulated in the 20 W mouse ovary. In this study, we aimed to identify key miRNAs and their target genes encompassing the reproductive span of mouse ovaries using mRNA and miRNA sequencing. These results indicated that gene sets are regulated in the reproductive stage-specific manner via interaction with miRNAs. Furthermore, consistent expression of Ccnd2 and Igf1 is considered crucial for the ovarian reserve and is regulated by many interactive miRNAs.
Collapse
Affiliation(s)
- Yoon-Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-W.K.); (H.K.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Kwang-Soo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul 03080, Korea;
| | - Yong-Jin Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Goryeodae-ro 73, Seongbuk-gu, Seoul 02841, Korea;
| | - Sung-Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-W.K.); (H.K.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-W.K.); (H.K.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-W.K.); (H.K.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
17
|
Mesa AM, Mao J, Medrano TI, Bivens NJ, Jurkevich A, Tuteja G, Cooke PS, Rosenfeld CS. Spatial Transcriptomics analysis of uterine gene expression in enhancer of Zeste homolog 2 (Ezh2) conditional knockout mice. Biol Reprod 2021; 105:1126-1139. [PMID: 34344022 DOI: 10.1093/biolre/ioab147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Histone proteins undergo various modifications that alter chromatin structure, including addition of methyl groups. Enhancer of homolog 2 (EZH2), is a histone methyltransferase that methylates lysine residue 27, and thereby, suppresses gene expression. EZH2 plays integral role in the uterus and other reproductive organs. We have previously shown that conditional deletion of uterine EZH2 results in increased proliferation of luminal and glandular epithelial cells, and RNAseq analyses reveal several uterine transcriptomic changes in Ezh2 conditional (c) knockout (KO) mice that can affect estrogen signaling pathways. To pinpoint the origin of such gene expression changes, we used the recently developed spatial transcriptomics (ST) method with the hypotheses that Ezh2cKO mice would predominantly demonstrate changes in epithelial cells and/or ablation of this gene would disrupt normal epithelial/stromal gene expression patterns. Uteri were collected from ovariectomized adult WT and Ezh2cKO mice and analyzed by ST. Asb4, Cxcl14, Dio2, and Igfbp5 were increased, Sult1d1, Mt3, and Lcn2 were reduced in Ezh2cKO uterine epithelium vs. WT epithelium. For Ezh2cKO uterine stroma, differentially expressed key hub genes included Cald1, Fbln1, Myh11, Acta2, and Tagln. Conditional loss of uterine Ezh2 also appears to shift the balance of gene expression profiles in epithelial vs. stromal tissue toward uterine epithelial cell and gland development and proliferation, consistent with uterine gland hyperplasia in these mice. Current findings provide further insight into how EZH2 may selectively affect uterine epithelial and stromal compartments. Additionally, these transcriptome data might provide the mechanistic understanding and valuable biomarkers for human endometrial disorders with epigenetic underpinnings.
Collapse
Affiliation(s)
- Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA.,Grupo de Investigación en Agrociencias, Biodiversidad y Territorio - GAMMA, Facultad de Ciencias Agrarias, Universidad de Antioquia UdeA, Calle 70 N° 52-21, Medellín, Colombia
| | - Jiude Mao
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Nathan J Bivens
- Genomics Technology, University of Missouri, Columbia, MO 65211, USA
| | - Alexander Jurkevich
- Advanced Light Microscopy Core Facility, University of Missouri, Columbia, MO 65211, USA
| | - Geetu Tuteja
- Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Cheryl S Rosenfeld
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA.,Data Science and Informatics Institute, University of Missouri, Columbia; MO 65211, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
18
|
Cooke PS, Mesa AM, Sirohi VK, Levin ER. Role of nuclear and membrane estrogen signaling pathways in the male and female reproductive tract. Differentiation 2020; 118:24-33. [PMID: 33339644 DOI: 10.1016/j.diff.2020.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Estrogen signaling through the main estrogen receptor, estrogen receptor 1 (ESR1; also known as ERα), is essential for normal female and male reproductive function. Historically, studies of estrogen action have focused on the classical genomic pathway. Although this is clearly the major pathway for steroid hormone actions, these hormones also signal through rapid non-classical effects involving cell membrane actions. Reports of rapid effects of estrogens extend for more than half a century, but recent results have expanded understanding of the identity, structure, function and overall importance of membrane receptors in estrogen responses. Key findings in this field were the immunohistochemical detection of ESR1 in cell membranes and demonstration that a portion of newly synthesized ESR1 is routed to the membrane by palmitoylation. These receptors in the membrane can then signal through protein kinases and other mechanisms following ligand binding to alter cell function. Another crucial advance in the field was development of transgenic mice expressing normal amounts of functional nuclear ESR1 (nESR1) but lacking membrane ESR1 (mESR1). Both male and female transgenic mice lacking mESR1 were infertile as adults, and both sexes had extensive reproductive abnormalities. Transgenic mice lacking mESR1 were highly protected from deleterious effects of neonatal estrogen administration, and estrogen effects on the histone methyltransferase Enhancer of Zeste homolog 2 that are mediated through mESR1 could have significant effects on epigenetic imprinting. In summary, signaling through mESR1 is essential for normal male and female reproductive function and fertility, and is a critical enabler of normal estrogen responses in vivo. Although the precise role of mESR1 in estrogen responses remains to be established, future research in this area should clarify its mechanism of action and lead to a better understanding of how mESR1 signaling works with classical genomic signaling through nESR1 to promote full estrogenic responses.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA.
| | - Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA
| | - Vijay K Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA; Department of Veterans Affairs Medical Center, Long Beach, Long Beach, CA, 90822, USA
| |
Collapse
|
19
|
Fang X, Ni N, Gao Y, Lydon JP, Ivanov I, Rijnkels M, Bayless KJ, Li Q. Transforming growth factor beta signaling and decidual integrity in mice†. Biol Reprod 2020; 103:1186-1198. [PMID: 32902612 PMCID: PMC7711917 DOI: 10.1093/biolre/ioaa155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/28/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor beta (TGFβ) signaling regulates multifaceted reproductive processes. It has been shown that the type 1 receptor of TGFβ (TGFBR1) is indispensable for female reproductive tract development, implantation, placental development, and fertility. However, the role of TGFβ signaling in decidual development and function remains poorly defined. Our objective is to determine the impact of uterine-specific deletion of Tgfbr1 on decidual integrity, with a focus on the cellular and molecular properties of the decidua during development. Our results show that the developmental dynamics of the decidua is altered in TGFBR1 conditionally depleted uteri from embryonic day (E) 5.5 to E8.5, substantiated by downregulation of genes associated with inflammatory responses and uterine natural killer cell abundance, reduced presence of nondecidualized fibroblasts in the antimesometrial region, and altered decidual cell development. Notably, conditional ablation of TGFBR1 results in the formation of decidua containing more abundant alpha smooth muscle actin (ACTA2)-positive cells at the peripheral region of the antimesometrial side versus controls at E6.5-E8.5. This finding is corroborated by upregulation of a subset of smooth muscle marker genes in Tgfbr1 conditionally deleted decidua at E6.5 and E8.5. Moreover, increased cell proliferation and enhanced decidual ERK1/2 signaling were found in Tgfbr1 conditional knockout mice upon decidual regression. In summary, conditional ablation of TGFBR1 in the uterus profoundly impacts the cellular and molecular properties of the decidua. Our results suggest that TGFBR1 in uterine epithelial and stromal compartments is important for the integrity of the decidua, a transient but crucial structure that supports embryo development.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Yang Gao
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Monique Rijnkels
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
20
|
Abstract
Epigenetic modifications regulate normal physiological, as well as pathological processes in various organs, including the uterus and placenta. Both organs undergo dramatic and rapid restructuring that depends upon precise orchestration of events. Epigenetic changes that alter transcription and translation of gene-sets regulate such responses. Histone modifications alter the chromatin structure, thereby affecting transcription factor access to gene promoter regions. Binding of histones to DNA is regulated by addition or removal of subunit methyl and other groups, which can inhibit or stimulate transcription. Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of polycomb repressive complex 2 (PRC2) that catalyzes tri-methylation of histone H3 at Lys 27 (H3K27me3) and subsequently suppresses transcription of genes bound by such histones. Uterine EZH2 expression exerts a critical role in development and function of this organ with deletion of this gene resulting in uterine hyperplasia and expression of cancer-associated transcripts. Elucidating the roles of EZH2 in uterus and placenta is essential as EZH2 dysregulation is associated with several uterine and placental pathologies. Herein, we discuss EZH2 functions in uterus and placenta, emphasizing its physiological and pathological importance.
Collapse
|
21
|
Hewitt SC, Carmona M, Foley KG, Donoghue LJ, Lierz SL, Winuthayanon W, Korach KS. Peri- and Postpubertal Estrogen Exposures of Female Mice Optimize Uterine Responses Later in Life. Endocrinology 2020; 161:bqaa081. [PMID: 32623449 PMCID: PMC7417879 DOI: 10.1210/endocr/bqaa081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/14/2020] [Indexed: 01/12/2023]
Abstract
At birth, all female mice, including those that either lack estrogen receptor α (ERα-knockout) or that express mutated forms of ERα (AF2ERKI), have a hypoplastic uterus. However, uterine growth and development that normally accompany pubertal maturation does not occur in ERα-knockout or AF2ERKI mice, indicating ERα-mediated estrogen (E2) signaling is essential for this process. Mice that lack Cyp19 (aromatase knockout, ArKO mice), an enzyme critical for E2 synthesis, are unable to make E2 and lack pubertal uterine development. A single injection of E2 into ovariectomized adult (10 weeks old) females normally results in uterine epithelial cell proliferation; however, we observe that although ERα is present in the ArKO uterine cells, no proliferative response is seen. We assessed the impact of exposing ArKO mice to E2 during pubertal and postpubertal windows and observed that E2-exposed ArKO mice acquired growth responsiveness. Analysis of differential gene expression between unexposed ArKO samples and samples from animals exhibiting the ability to mount an E2-induced uterine growth response (wild-type [WT] or E2-exposed ArKO) revealed activation of enhancer of zeste homolog 2 (EZH2) and heart- and neural crest derivatives-expressed protein 2 (HAND2) signaling and inhibition of GLI Family Zinc Finger 1 (GLI1) responses. EZH2 and HAND2 are known to inhibit uterine growth, and GLI1 is involved in Indian hedgehog signaling, which is a positive mediator of uterine response. Finally, we show that exposure of ArKO females to dietary phytoestrogens results in their acquisition of uterine growth competence. Altogether, our findings suggest that pubertal levels of endogenous and exogenous estrogens impact biological function of uterine cells later in life via ERα-dependent mechanisms.
Collapse
Affiliation(s)
- Sylvia C Hewitt
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Marleny Carmona
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - K Grace Foley
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Lauren J Donoghue
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Sydney L Lierz
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Wipawee Winuthayanon
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Kenneth S Korach
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| |
Collapse
|
22
|
Mesa AM, Mao J, Nanjappa MK, Medrano TI, Tevosian S, Yu F, Kinkade J, Lyu Z, Liu Y, Joshi T, Wang D, Rosenfeld CS, Cooke PS. Mice lacking uterine enhancer of zeste homolog 2 have transcriptomic changes associated with uterine epithelial proliferation. Physiol Genomics 2020; 52:81-95. [PMID: 31841397 PMCID: PMC7052568 DOI: 10.1152/physiolgenomics.00098.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 01/16/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that suppresses gene expression. Previously, we developed a conditional null model where EZH2 is knocked out in uterus. Deletion of uterine EZH2 increased proliferation of luminal and glandular epithelial cells. Herein, we used RNA-Seq in wild-type (WT) and EZH2 conditional knockout (Ezh2cKO) uteri to obtain mechanistic insights into the gene expression changes that underpin the pathogenesis observed in these mice. Ovariectomized adult Ezh2cKO mice were treated with vehicle (V) or 17β-estradiol (E2; 1 ng/g). Uteri were collected at postnatal day (PND) 75 for RNA-Seq or immunostaining for epithelial proliferation. Weighted gene coexpression network analysis was used to link uterine gene expression patterns and epithelial proliferation. In V-treated mice, 88 transcripts were differentially expressed (DEG) in Ezh2cKO mice, and Bmp5, Crabp2, Lgr5, and Sprr2f were upregulated. E2 treatment resulted in 40 DEG with Krt5, Krt15, Olig3, Crabp1, and Serpinb7 upregulated in Ezh2cKO compared with control mice. Transcript analysis relative to proliferation rates revealed two module eigengenes correlated with epithelial proliferation in WT V vs. Ezh2cKO V and WT E2 vs. Ezh2cKO E2 mice, with a positive relationship in the former and inverse in the latter. Notably, the ESR1, Wnt, and Hippo signaling pathways were among those functionally enriched in Ezh2cKO females. Current results reveal unique gene expression patterns in Ezh2cKO uterus and provide insight into how loss of this critical epigenetic regulator assumingly contributes to uterine abnormalities.
Collapse
Affiliation(s)
- Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
- Grupo de Investigación en Génetica, Mejoramiento y Modelación Animal-GaMMA, Universidad de Antioquia, Medellín, Colombia
| | - Jiude Mao
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
| | | | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| | - Sergei Tevosian
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| | - Fahong Yu
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Missouri
| | - Jessica Kinkade
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Zhen Lyu
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
| | - Yang Liu
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Informatics Institute, University of Missouri, Columbia; Missouri
| | - Trupti Joshi
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
- Informatics Institute, University of Missouri, Columbia; Missouri
- Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, Missouri
| | - Duolin Wang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri
| | - Cheryl S Rosenfeld
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Biomedical Sciences, University of Missouri, Columbia, Missouri
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Missouri
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
23
|
Kong S, Zhou C, Bao H, Ni Z, Liu M, He B, Huang L, Sun Y, Wang H, Lu J. Epigenetic control of embryo-uterine crosstalk at peri-implantation. Cell Mol Life Sci 2019; 76:4813-4828. [PMID: 31352535 PMCID: PMC11105790 DOI: 10.1007/s00018-019-03245-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 01/05/2023]
Abstract
Embryo implantation is one of the pivotal steps during mammalian pregnancy, since the quality of embryo implantation determines the outcome of ongoing pregnancy and fetal development. A large number of factors, including transcription factors, signalling transduction components, and lipids, have been shown to be indispensable for embryo implantation. Increasing evidence also suggests the important roles of epigenetic factors in this critical event. This review focuses on recent findings about the involvement of epigenetic regulators during embryo implantation.
Collapse
Affiliation(s)
- Shuangbo Kong
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Chan Zhou
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Zhangli Ni
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Mengying Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Bo He
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Lin Huang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Yang Sun
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Haibin Wang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| | - Jinhua Lu
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| |
Collapse
|
24
|
Wang X, Hawkins SM. Uterine Epithelial Development and Enhancer of Zeste Homolog 2: It Is Important for More than Just Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1176-1177. [PMID: 30986382 DOI: 10.1016/j.ajpath.2019.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 10/27/2022]
Abstract
This commentary highlights the article by Fang et al that describes the role of enhancer of zeste homolog 2 in endometrial development.
Collapse
Affiliation(s)
- Xiyin Wang
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shannon M Hawkins
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|