1
|
Xypolita ME, Goolam M, Bikoff EK, Robertson EJ, Mould AW. The zinc-finger transcription factor Blimp1/Prdm1 is required for uterine remodelling and repair in the mouse. Nat Commun 2025; 16:1220. [PMID: 39890816 PMCID: PMC11785775 DOI: 10.1038/s41467-025-56511-8] [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: 12/18/2023] [Accepted: 01/17/2025] [Indexed: 02/03/2025] Open
Abstract
The zinc finger transcription factor Blimp1/PRDM1 regulates gene expression in diverse cell types. Its activity controls the maternal decidual response at early post-implantation stages of development. The present experiments demonstrate surprisingly that Blimp1 activity in the uterus is required for tissue remodelling at sites of embryonic failure. Moreover Blimp1 mutant females are refractory to RU486 induced decidual shedding. RNA-seq together with immunostaining experiments strongly suggest that the failure to up-regulate expression of the matrix metalloprotease Mmp10 in combination with insufficient suppression of BMP signalling, likely explain Blimp1-dependent phenotypic changes. In the post-partum uterus Blimp1 together with Mmp10 are highly upregulated at sites of tissue repair following placental detachment. Conditional Blimp1 removal significantly impairs the re-epithelization process and severely impacts involution of the endometrium and luminal epithelium. Overall these results identify Blimp1 as a master regulator of uterine tissue remodelling and repair.
Collapse
Affiliation(s)
- Maria-Eleni Xypolita
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Mubeen Goolam
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Human Biology and Neuroscience Institute, University of Cape Town, Cape Town, 7925, South Africa
| | - Elizabeth K Bikoff
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Elizabeth J Robertson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Arne W Mould
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
2
|
Roberson EC, Tran NK, Godambe AN, Mark H, Nguimtsop M, Rust T, Ung E, Barker LJ, Fitch RD, Wallingford JB. Hedgehog signaling is required for endometrial remodeling and myometrial homeostasis in the cycling mouse uterus. iScience 2023; 26:107993. [PMID: 37810243 PMCID: PMC10551904 DOI: 10.1016/j.isci.2023.107993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/24/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023] Open
Abstract
Decades of work demonstrate that the mammalian estrous cycle is controlled by cycling steroid hormones. However, the signaling mechanisms that act downstream, linking hormonal action to the physical remodeling of the cycling uterus, remain unclear. To address this issue, we analyzed gene expression at all stages of the mouse estrous cycle. Strikingly, we found that several genetic programs well-known to control tissue morphogenesis in developing embryos displayed cyclical patterns of expression. We find that most of the genetic architectures of Hedgehog signaling (ligands, receptors, effectors, and transcription factors) are transcribed cyclically in the uterus, and that conditional disruption of the Hedgehog receptor smoothened not only elicits a failure of normal cyclical thickening of the endometrial lining but also induces aberrant deformation of the uterine smooth muscle. Together, our data shed light on the mechanisms underlying normal uterine remodeling specifically and cyclical gene expression generally.
Collapse
Affiliation(s)
- Elle C. Roberson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Ngan Kim Tran
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Anushka N. Godambe
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Harrison Mark
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michelle Nguimtsop
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Trinity Rust
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Elizabeth Ung
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - LeCaine J. Barker
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Rebecca D. Fitch
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
3
|
He W, Zhu X, Xin A, Zhang H, Sun Y, Xu H, Li H, Yang T, Zhou D, Yan H, Sun X. Long-term maintenance of human endometrial epithelial stem cells and their therapeutic effects on intrauterine adhesion. Cell Biosci 2022; 12:175. [PMID: 36258228 PMCID: PMC9580151 DOI: 10.1186/s13578-022-00905-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/25/2022] [Indexed: 11/05/2022] Open
Abstract
Background The human endometrium is a highly regenerative tissue that is believed to have two main types of stem cells: endometrial mesenchymal/stromal stem cells (eMSCs) and endometrial epithelial stem cells (eESCs). So far, eMSCs have been extensively studied, whereas the studies of eESCs are constrained by the inability to culture and expand them in vitro. The aim of this study is to establish an efficient method for the production of eESCs from human endometrium for potential clinical application in intrauterine adhesion (IUA). Results Here we developed a culture condition with a combination of some small molecules for in vitro culturing and expansion of human SSEA-1+ cells. The SSEA-1+ cells exhibited stem/progenitor cell activity in vitro, including clonogenicity and differentiation capacity into endometrial epithelial cell-like cells. In addition, the SSEA-1+ cells, embedded in extracellular matrix, swiftly self-organized into organoid structures with long-term expansion capacity and histological phenotype of the human endometrial epithelium. Specifically, we found that the SSEA-1+ cells showed stronger therapeutic potential than eMSCs for IUA in vitro. In a rat model of IUA, in situ injection of the SSEA-1+ cells-laden chitosan could efficiently reduce fibrosis and facilitate endometrial regeneration. Conclusions Our work demonstrates an approach for isolation and expansion of human eESCs in vitro, and an appropriate marker, SSEA-1, to identify eESCs. Furthermore, the SSEA-1+ cells-laden chitosan might provide a novel cell-based approach for IUA treatment. These findings will advance the understanding of pathophysiology during endometrial restoration which may ultimately lead to more rational clinical practice. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00905-4.
Collapse
Affiliation(s)
- Wen He
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xuejing Zhu
- Shanghai Celliver Biotechnology Co. Ltd, Shanghai, China
| | - Aijie Xin
- grid.8547.e0000 0001 0125 2443NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Hongdan Zhang
- Shanghai Celliver Biotechnology Co. Ltd, Shanghai, China
| | - Yiming Sun
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Hua Xu
- grid.412312.70000 0004 1755 1415Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - He Li
- grid.412312.70000 0004 1755 1415Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Tianying Yang
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Dan Zhou
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Hexin Yan
- Shanghai Celliver Biotechnology Co. Ltd, Shanghai, China
| | - Xiaoxi Sun
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China ,grid.412312.70000 0004 1755 1415Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China ,grid.412312.70000 0004 1755 1415Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
4
|
Zhang S, Chan RWS, Ng EHY, Yeung WSB. The role of Notch signaling in endometrial mesenchymal stromal/stem-like cells maintenance. Commun Biol 2022; 5:1064. [PMID: 36207605 PMCID: PMC9547015 DOI: 10.1038/s42003-022-04044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/27/2022] [Indexed: 11/30/2022] Open
Abstract
Human endometrium undergoes cycles of regeneration in women of reproductive age. The endometrial mesenchymal stromal/stem cells (eMSC) contribute to this process. Notch signaling is essential for homeostasis of somatic stem cells. However, its role in eMSC remains unclear. We show with gain- and loss-of-function experiments that activation of Notch signaling promotes eMSC maintenance, while inhibition induces opposite effect. The activation of Notch signaling better maintains eMSC in a quiescent state. However, these quiescent eMSC can re-enter the cell cycle depending on the Notch and Wnt activities in the microenvironment, suggesting a crosstalk between the two signaling pathways. We further show that the Notch signaling is involved in endometrial remodeling event in a mouse menstrual-like model. Suppression of Notch signaling reduces the proliferation of Notch1+ label-retaining stromal cells and delays endometrial repair. Our data demonstrate the importance of Notch signaling in regulating the endometrial stem/progenitor cells in vitro and in vivo. Notch signaling promotes the quiescent state of endometrial mesenchymal stromal/stem cells (eMSC), whose re-rentry into the cell cycle is in turn influenced by Notch and Wnt signaling from the microenvironment.
Collapse
Affiliation(s)
- Sisi Zhang
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China.,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, 518000, China
| | - Rachel W S Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China. .,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, 518000, China.
| | - Ernest H Y Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China.,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, 518000, China
| | - William S B Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China. .,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, 518000, China.
| |
Collapse
|
5
|
Thieffry C, Van Wynendaele M, Samain L, Tyteca D, Pierreux C, Marbaix E, Henriet P. Spatiotemporal expression pattern of Progesterone Receptor Component (PGRMC) 1 in endometrium from patients with or without endometriosis or adenomyosis. J Steroid Biochem Mol Biol 2022; 223:106153. [PMID: 35835350 DOI: 10.1016/j.jsbmb.2022.106153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/15/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
The endometrium plays a crucial role in reproduction and, in humans, is cyclically remodeled under hormonal control. Estradiol favors tissue proliferation whereas progesterone inhibits tissue growth and induces morphological changes. Endometriosis is often associated with fertility issues and with exacerbated estrogen and reduced progesterone concentration or response in the eutopic endometrium. However, underlying mechanisms remain unclear. Progesterone Receptor Membrane Component (PGRMC) 1 is a protein able to modulate progesterone response and its murine knockout reduced fertility. However, the precise spatiotemporal pattern of PGRMC1 expression in the human endometrium is still poorly characterized. We investigated variations of eutopic endometrial PGRMC1 expression by combining RT-qPCR, immunofluorescence and in situ hybridization. We found that PGRMC1 expression progressively increases during the proliferative phase and decreases during the secretory phase. However, immunolabeling and identification of mRNA-containing cells were regularly heterogeneous in samples, according to tissue depth, with a gradient extending from the surface epithelium towards the basalis. There was no significant difference in PGRMC1 mRNA amounts between patients with or without endometriosis or adenomyosis, for any phase of the menstrual cycle, but cells with strong or moderate PGRMC1 immunolabeling were reduced during the proliferative phase in endometriotic patients. In conclusion, although the cyclical variation of PGRMC1 expression globally follows fluctuation of ovarian steroids, further work is required to precisely characterize hormonal control and identify the additional levels of regulation responsible for local adjustment of PGRMC1 concentration. This is particularly important in the light of recent studies emphasizing the correlation between adequate PGRMC1 amounts and fertility.
Collapse
Affiliation(s)
- Charlotte Thieffry
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| | - Marie Van Wynendaele
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| | - Lucie Samain
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| | - Donatienne Tyteca
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| | - Christophe Pierreux
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| | - Etienne Marbaix
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium; Pathology Department, Cliniques Universitaires Saint-Luc, B-1200 Brussels, Belgium.
| | - Patrick Henriet
- CELL Unit, De Duve Institute and Université Catholique de Louvain, B-1200 Brussels, Belgium.
| |
Collapse
|
6
|
Liu R, Dai M, Gong G, Chen M, Cao C, Wang T, Hou Z, Shi Y, Guo J, Zhang Y, Xia X. The role of extracellular matrix on unfavorable maternal–fetal interface: focusing on the function of collagen in human fertility. JOURNAL OF LEATHER SCIENCE AND ENGINEERING 2022. [DOI: 10.1186/s42825-022-00087-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AbstractExtracellular matrix (ECM) is characterized as widespread, abundant, and pluripotent. Among ECM members, collagen is widely accepted as one of the most prominent components for its essential structural property that can provide a scaffold for other components of ECM and the rich biological functions, which has been extensively used in tissue engineering. Emerging evidence has shown that the balance of ECM degradation and remodeling is vital to regulations of maternal–fetal interface including menstrual cycling, decidualization, embryo implantation and pregnancy maintenance. Moreover, disorders in these events may eventually lead to failure of pregnancy. Although the improvement of assisted conception and embryo culture technologies bring hope to many infertile couples, some unfavorable outcomes, such as recurrent implantation failure (RIF), recurrent pregnancy loss (RPL) or recurrent miscarriage (RM), keep troubling the clinicians and patients. Recently, in vitro three-dimensional (3D) model mimicking the microenvironment of the maternal–fetal interface is developed to investigate the physiological and pathological conditions of conception and pregnancy. The progress of this technology is based on clarifying the role of ECM in the endometrium and the interaction between endometrium and conceptus. Focusing on collagen, the present review summarized the degradation and regulation of ECM and its role in normal menstruation, endometrium receptivity and unsatisfying events occurring in infertility treatments, as well as the application in therapeutic approaches to improve pregnancy outcomes. More investigations about ECM focusing on the maternal–fetal interface interaction with mesenchymal stem cells or local immunoregulation may inspire new thoughts and advancements in the clinical application of infertility treatments.
Graphical abstract
Collapse
|
7
|
Kirkwood PM, Shaw IW, Saunders PTK. Mechanisms of Scarless Repair at Time of Menstruation: Insights From Mouse Models. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 3:801843. [PMID: 36304046 PMCID: PMC9580659 DOI: 10.3389/frph.2021.801843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022] Open
Abstract
The human endometrium is a remarkable tissue which may experience up to 400 cycles of hormone-driven proliferation, differentiation and breakdown during a woman's reproductive lifetime. During menstruation, when the luminal portion of tissue breaks down, it resembles a bloody wound with piecemeal shedding, exposure of underlying stroma and a strong inflammatory reaction. In the absence of pathology within a few days the integrity of the tissue is restored without formation of a scar and the endometrium is able to respond appropriately to subsequent endocrine signals in preparation for establishment of pregnancy if fertilization occurs. Understanding mechanisms regulating scarless repair of the endometrium is important both for design of therapies which can treat conditions where this is aberrant (heavy menstrual bleeding, fibroids, endometriosis, Asherman's syndrome) as well as to provide new information that might allow us to reduce fibrosis and scar formation in other tissues. Menstruation only occurs naturally in species that exhibit spontaneous stromal cell decidualization during the fertile cycle such as primates (including women) and the Spiny mouse. To take advantage of genetic models and detailed time course analysis, mouse models of endometrial shedding/repair involving hormonal manipulation, artificial induction of decidualization and hormone withdrawal have been developed and refined. These models are useful in modeling dynamic changes across the time course of repair and have recapitulated key features of endometrial repair in women including local hypoxia and immune cell recruitment. In this review we will consider the evidence that scarless repair of endometrial tissue involves changes in stromal cell function including mesenchyme to epithelial transition, epithelial cell proliferation and multiple populations of immune cells. Processes contributing to endometrial fibrosis (Asherman's syndrome) as well as scarless repair of other tissues including skin and oral mucosa are compared to that of menstrual repair.
Collapse
|
8
|
Sanchez-Mata A, Gonzalez-Muñoz E. Understanding menstrual blood-derived stromal/stem cells: Definition and properties. Are we rushing into their therapeutic applications? iScience 2021; 24:103501. [PMID: 34917895 PMCID: PMC8646170 DOI: 10.1016/j.isci.2021.103501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cells with mesenchymal stem cell properties have been identified in menstrual blood and termed menstrual blood-derived stem/stromal cells (MenSCs). MenSCs have been proposed as ideal candidates for cell-based therapy in regenerative medicine and immune-related diseases. However, MenSCs identity has been loosely defined so far and there is controversy regarding their cell markers and differentiation potential. In this review, we outline the origin of MenSCs in the context of regenerating human endometrium, with attention to endometrial eMSCs as reference cells to understand MenSCs. We summarize the cell identity markers analyzed and the immunomodulatory and reparative properties reported. We also address the recent use of MenSCs in cell reprogramming. The main goal of this review is to contribute to the understanding of the identity and properties of MenSCs as well as to identify potential caveats and new venues that deserve to be explored to strengthen their potential applications.
Collapse
Affiliation(s)
- Alicia Sanchez-Mata
- Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29071 Málaga, Spain
| | - Elena Gonzalez-Muñoz
- Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29071 Málaga, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), 29071 Málaga, Spain
| |
Collapse
|
9
|
Kolanska K, Sbeih M, Canlorbe G, Mekinian A, Varinot J, Capmas P, Koskas M, Aractingi S, Daraï E, Chabbert-Buffet N. Ulipristal Acetate Modifies miRNA Expression in Both Superficial and Basal Layers of the Human Endometrium. J Clin Med 2021; 10:jcm10194442. [PMID: 34640460 PMCID: PMC8509688 DOI: 10.3390/jcm10194442] [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: 09/04/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/05/2022] Open
Abstract
(1) Background: Ulipristal acetate (UPA) is a selective progesterone receptor modulator (SPRM) widely used for emergency contraception and mid- to long-term leiomyoma treatment. The aim of this study was to identify modifications of miRNA expression in superficial and basal layers of the human endometrium at the end of the UPA treatment for at least 3 months. (2) Methods: Microarray miRNA analysis of formalin-fixed, paraffin-embedded hysterectomy tissue samples was conducted, followed by an Ingenuity Pathway Analysis. Samples were divided into three groups: women having had 3 months of UPA treatment (n = 7); and two control groups of UPA-naïve women in the proliferative (n = 8) or secretory (n = 6) phase. (3) Results: The UPA modified the expression of 59 miRNAs involved in the processes of cell cycle, carcinogenesis, and inflammation. Their expression profiles were different in the basal and superficial layers. Most of the processes influenced by the UPA in the basal layer were connected to the cell cycle and immune regulation. (4) Conclusion: Specific changes were observed in both layers of the endometrium in the UPA group. However, the miRNA expression in the basal layer was not consistent with that in the superficial layer. Other large studies analysing the long-term impact of SPRM on endometrial miRNA expression are necessary.
Collapse
Affiliation(s)
- Kamila Kolanska
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
- Service de Gynécologie Sestertius et Médecine de la Reproduction, AP-HP Sorbonne Université Site Tenon, 4 rue de la Chine, 75020 Paris, France
- Correspondence:
| | - Maria Sbeih
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
| | - Geoffroy Canlorbe
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
- Department of Gynecological and Breast Surgery and Oncology, Pitié-Salpêtrière University Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), 75013 Paris, France
| | - Arsène Mekinian
- Service de Médecine Interne, AP-HP Sorbonne Université Site St Antoine, 184 rue du Faubourg Saint Antoine, 75012 Paris, France;
| | - Justine Varinot
- Service d’Anatomopathologie, AP HP Sorbonne Université Site Tenon, 4 rue de la Chine, 75020 Paris, France;
| | - Perrine Capmas
- Department of Gynecology and Obstetrics, University Paris Saclay, 78 rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France;
- Inserm, Centre of Research in Epidemiology and Population Health (CESP), U1018, 94276 Le Kremlin-Bicêtre, France
| | - Martin Koskas
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
- Department of Obstetrics and Gynecology, AP-HP Bichat University Hospital, 75018 Paris, France
- Institut de Recherche en Santé de la Femme, Equipe d’accueil 7285, Universite de Versailles Saint-Quentin-en-Yvelines, 78180 Montigny-le-Bretonneux, France
| | - Selim Aractingi
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
| | - Emile Daraï
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
- Service de Gynécologie Sestertius et Médecine de la Reproduction, AP-HP Sorbonne Université Site Tenon, 4 rue de la Chine, 75020 Paris, France
| | - Nathalie Chabbert-Buffet
- INSERM UMRS 938, Sorbonne Université, Site Saint-Antoine, 27 rue Chaligny, CEDEX 12, 75571 Paris, France; (M.S.); (G.C.); (M.K.); (S.A.); (E.D.); (N.C.-B.)
- Service de Gynécologie Sestertius et Médecine de la Reproduction, AP-HP Sorbonne Université Site Tenon, 4 rue de la Chine, 75020 Paris, France
| |
Collapse
|
10
|
Abstract
Uniquely among adult tissues, the human endometrium undergoes cyclical shedding, scar-free repair and regeneration during a woman's reproductive life. Therefore, it presents an outstanding model for study of such processes. This Review examines what is known of endometrial repair and regeneration following menstruation and parturition, including comparisons with wound repair and the influence of menstrual fluid components. We also discuss the contribution of endometrial stem/progenitor cells to endometrial regeneration, including the importance of the stem cell niche and stem cell-derived extracellular vesicles. Finally, we comment on the value of endometrial epithelial organoids to extend our understanding of endometrial development and regeneration, as well as therapeutic applications.
Collapse
Affiliation(s)
- Lois A Salamonsen
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Jennifer C Hutchison
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Caroline E Gargett
- Ritchie Centre, Hudson Institute of Medical Research, 25-31 Wright St, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
11
|
Cousins FL, Pandoy R, Jin S, Gargett CE. The Elusive Endometrial Epithelial Stem/Progenitor Cells. Front Cell Dev Biol 2021; 9:640319. [PMID: 33898428 PMCID: PMC8063057 DOI: 10.3389/fcell.2021.640319] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
The human endometrium undergoes approximately 450 cycles of proliferation, differentiation, shedding and regeneration over a woman's reproductive lifetime. The regenerative capacity of the endometrium is attributed to stem/progenitor cells residing in the basalis layer of the tissue. Mesenchymal stem cells have been extensively studied in the endometrium, whereas endometrial epithelial stem/progenitor cells have remained more elusive. This review details the discovery of human and mouse endometrial epithelial stem/progenitor cells. It highlights recent significant developments identifying putative markers of these epithelial stem/progenitor cells that reveal their in vivo identity, location in both human and mouse endometrium, raising common but also different viewpoints. The review also outlines the techniques used to identify epithelial stem/progenitor cells, specifically in vitro functional assays and in vivo lineage tracing. We will also discuss their known interactions and hierarchy and known roles in endometrial dynamics across the menstrual or estrous cycle including re-epithelialization at menses and regeneration of the tissue during the proliferative phase. We also detail their potential role in endometrial proliferative disorders such as endometriosis.
Collapse
Affiliation(s)
- Fiona L. Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynecology, Monash University, Clayton, VIC, Australia
| | - Ronald Pandoy
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Shiying Jin
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
12
|
Reavey JJ, Walker C, Nicol M, Murray AA, Critchley HOD, Kershaw LE, Maybin JA. Markers of human endometrial hypoxia can be detected in vivo and ex vivo during physiological menstruation. Hum Reprod 2021; 36:941-950. [PMID: 33496337 PMCID: PMC7970728 DOI: 10.1093/humrep/deaa379] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/17/2020] [Indexed: 12/23/2022] Open
Abstract
STUDY QUESTION Can markers of human endometrial hypoxia be detected at menstruation in vivo? SUMMARY ANSWER Our in vivo data support the presence of hypoxia in menstrual endometrium of women during physiological menstruation. WHAT IS KNOWN ALREADY Current evidence from animal models and human in vitro studies suggests endometrial hypoxia is present at menstruation and drives endometrial repair post menses. However, detection of human endometrial hypoxia in vivo remains elusive. STUDY DESIGN, SIZE, DURATION We performed a prospective case study of 16 women with normal menstrual bleeding. PARTICIPANTS/MATERIALS, SETTING, METHODS Reproductively aged female participants with a regular menstrual cycle underwent objective measurement of their menstrual blood loss using the alkaline haematin method to confirm a loss of <80 ml per cycle. Exclusion criteria were exogenous hormone use, an intrauterine device, endometriosis or fibroids >3 cm. Participants attended for two MRI scans; during days 1-3 of menstruation and the early/mid-secretory phase of their cycle. The MRI protocol included dynamic contrast-enhanced MRI and T2* quantification. At each visit, an endometrial sample was also collected and hypoxia-regulated repair factor mRNA levels (ADM, VEGFA, CXCR4) were quantified by RT-qPCR. MAIN RESULTS AND THE ROLE OF CHANCE Women had reduced T2* during menstrual scans versus non-menstrual scans (P = 0.005), consistent with menstrual hypoxia. Plasma flow (Fp) was increased at menstruation compared to the non-menstrual phase (P = 0.0005). Laboratory findings revealed increased ADM, VEGF-A and CXCR4 at menstruation on examination of paired endometrial biopsies from the menstrual and non-menstrual phase (P = 0.008; P = 0.03; P = 0.009). There was a significant correlation between T2* and these ex vivo hypoxic markers (P < 0.05). LIMITATIONS, REASONS FOR CAUTION This study examined the in vivo detection of endometrial hypoxic markers at specific timepoints in the menstrual cycle in women with a menstrual blood loss <80 ml/cycle and without significant uterine structural abnormalities. Further research is required to determine the presence of endometrial hypoxia in those experiencing abnormal uterine bleeding with and without fibroids/adenomyosis. WIDER IMPLICATIONS OF THE FINDINGS Heavy menstrual bleeding (HMB) is a common, debilitating condition. Understanding menstrual physiology may improve therapeutics. To our knowledge, this is the first in vivo data supporting the presence of menstrual hypoxia in the endometrium of women with normal menstrual bleeding. If aberrant in those with HMB, these non-invasive tests may aid diagnosis and facilitate personalized treatments for HMB. STUDY FUNDING/COMPETING INTEREST(S) This work was funded by Wellbeing of Women grant RG1820, Wellcome Trust Fellowship 209589/Z/17/Z and undertaken in the MRC Centre for Reproductive Health, funded by grants G1002033 and MR/N022556/1. H.O.D.C. has clinical research support for laboratory consumables and staff from Bayer AG and provides consultancy advice (but with no personal remuneration) for Bayer AG, PregLem SA, Gedeon Richter, Vifor Pharma UK Ltd, AbbVie Inc; Myovant Sciences GmbH. H.O.D.C. receives royalties from UpToDate for articles on abnormal uterine bleeding. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- J J Reavey
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| | - C Walker
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| | - M Nicol
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A A Murray
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| | - H O D Critchley
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| | - L E Kershaw
- Edinburgh Imaging, The Queen’s Medical Research Institute, Edinburgh, UK
- Centre for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh, UK
| | - J A Maybin
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh, UK
| |
Collapse
|
13
|
Reske JJ, Wilson MR, Holladay J, Wegener M, Adams M, Chandler RL. SWI/SNF inactivation in the endometrial epithelium leads to loss of epithelial integrity. Hum Mol Genet 2020; 29:3412-3430. [PMID: 33075803 PMCID: PMC7749707 DOI: 10.1093/hmg/ddaa227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/01/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Although ARID1A mutations are a hallmark feature, mutations in other SWI/SNF (SWItch/Sucrose Non-Fermentable) chromatin remodeling subunits are also observed in endometrial neoplasms. Here, we interrogated the roles of Brahma/SWI2-related gene 1 (BRG1, SMARCA4), the SWI/SNF catalytic subunit, in the endometrial epithelium. BRG1 loss affects more than one-third of all active genes and highly overlaps with the ARID1A gene regulatory network. Chromatin immunoprecipitation studies revealed widespread subunit-specific differences in transcriptional regulation, as BRG1 promoter interactions are associated with gene activation, while ARID1A binding is associated with gene repression. However, we identified a physiologically relevant subset of BRG1 and ARID1A co-regulated epithelial identity genes. Mice were genetically engineered to inactivate BRG1 specifically in the endometrial epithelium. Endometrial glands were observed embedded in uterine myometrium, indicating adenomyosis-like phenotypes. Molecular similarities were observed between BRG1 and ARID1A mutant endometrial cells in vivo, including loss of epithelial cell adhesion and junction genes. Collectively, these studies illustrate overlapping contributions of multiple SWI/SNF subunit mutations in the translocation of endometrium to distal sites, with loss of cell integrity being a common feature in SWI/SNF mutant endometrial epithelia.
Collapse
Affiliation(s)
- Jake J Reske
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Mike R Wilson
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Jeanne Holladay
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Marc Wegener
- Genomics Core Facility, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Marie Adams
- Genomics Core Facility, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ronald L Chandler
- To whom correspondence should be addressed at: Grand Rapids Research Center, 400 Monroe NW, Grand Rapids, MI 49503, USA. Tel: +1 6162340980;
| |
Collapse
|
14
|
Yu HN, Li XM, Kong LL, Ren J, Wu H, Bu LG, Ding NZ, Ni H. Connective tissue growth factor gene expression in goat endometrium during estrous cycle and early pregnancy. Theriogenology 2020; 153:85-90. [DOI: 10.1016/j.theriogenology.2020.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 12/31/2022]
|
15
|
Critchley HOD, Maybin JA, Armstrong GM, Williams ARW. Physiology of the Endometrium and Regulation of Menstruation. Physiol Rev 2020; 100:1149-1179. [DOI: 10.1152/physrev.00031.2019] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The physiological functions of the uterine endometrium (uterine lining) are preparation for implantation, maintenance of pregnancy if implantation occurs, and menstruation in the absence of pregnancy. The endometrium thus plays a pivotal role in reproduction and continuation of our species. Menstruation is a steroid-regulated event, and there are alternatives for a progesterone-primed endometrium, i.e., pregnancy or menstruation. Progesterone withdrawal is the trigger for menstruation. The menstruating endometrium is a physiological example of an injured or “wounded” surface that is required to rapidly repair each month. The physiological events of menstruation and endometrial repair provide an accessible in vivo human model of inflammation and tissue repair. Progress in our understanding of endometrial pathophysiology has been facilitated by modern cellular and molecular discovery tools, along with animal models of simulated menses. Abnormal uterine bleeding (AUB), including heavy menstrual bleeding (HMB), imposes a massive burden on society, affecting one in four women of reproductive age. Understanding structural and nonstructural causes underpinning AUB is essential to optimize and provide precision in patient management. This is facilitated by careful classification of causes of bleeding. We highlight the crucial need for understanding mechanisms underpinning menstruation and its aberrations. The endometrium is a prime target tissue for selective progesterone receptor modulators (SPRMs). This class of compounds has therapeutic potential for the clinical unmet need of HMB. SPRMs reduce menstrual bleeding by mechanisms still largely unknown. Human menstruation remains a taboo topic, and many questions concerning endometrial physiology that pertain to menstrual bleeding are yet to be answered.
Collapse
Affiliation(s)
- Hilary O. D. Critchley
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Jacqueline A. Maybin
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Gregory M. Armstrong
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Alistair R. W. Williams
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
16
|
YÜKSEL H, ZAFER E. Endometrial Stem/Progenitor Cells. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2020. [DOI: 10.1007/s13669-020-00278-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Muruganandan S, Fan X, Dhal S, Nayak NR. Development of A 3D Tissue Slice Culture Model for the Study of Human Endometrial Repair and Regeneration. Biomolecules 2020; 10:biom10010136. [PMID: 31947662 PMCID: PMC7022976 DOI: 10.3390/biom10010136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/28/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human endometrium undergoes sequential phases of shedding of the upper functionalis zone during menstruation, followed by regeneration of the functionalis zone from the remaining basalis zone cells, and secretory differentiation under the influence of the ovarian steroid hormones estradiol (E2) and progesterone (P4). This massive tissue regeneration after menstruation is believed to arise from endometrial stromal and epithelial stem cells residing in the basal layer of the endometrium. Although many endometrial pathologies are thought to be associated with defects in these stem cells, studies on their identification and regulation are limited, primarily due to lack of easily accessible animal models, as these processes are unique to primates. Here we describe a robust new method to study endometrial regeneration and differentiation processes using human endometrial tissue slice cultures incorporating an air-liquid interface into a 3D matrix scaffold of type I collagen gel, allowing sustained tissue viability over three weeks. The 3D collagen gel-embedded endometrial tissue slices in a double-dish culture system responded to ovarian steroid hormones, mimicking the endometrial changes that occur in vivo during the menstrual cycle. These changes included the E2-induced upregulation of Ki-67, estrogen receptor (ER), and progesterone receptor (PR) in all endometrial compartments and were markedly suppressed by both P4 and E2 plus P4 treatments. There were also distinct changes in endometrial morphology after E2 and P4 treatments, including subnuclear vacuolation and luminal secretions in glands as well as decidualization of stromal cells, typical characteristics of a progestational endometrium in vivo. This long-term slice culture method provides a unique in vivo-like microenvironment for the study of human endometrial functions and remodeling during early pregnancy and experiments on stem cell populations involved in endometrial regeneration and remodeling. Furthermore, this model has the potential to enable studies on several endometrial diseases, including endometrial cancers and pregnancy complications associated with defects in endometrial remodeling.
Collapse
Affiliation(s)
- Shanmugam Muruganandan
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Harvard University, Boston, MA 02115, USA
| | - Xiujun Fan
- Laboratory of Reproductive Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| | - Sabita Dhal
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
| | - Nihar R. Nayak
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
| |
Collapse
|
18
|
Cousins FL, O DF, Ong YR, Breault DT, Deane JA, Gargett CE. Telomerase Reverse Transcriptase Expression in Mouse Endometrium During Reepithelialization and Regeneration in a Menses-Like Model. Stem Cells Dev 2018; 28:1-12. [PMID: 30358490 DOI: 10.1089/scd.2018.0133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The regenerative capacity of the endometrium has been attributed to resident stem/progenitor cells. A number of stem/progenitor markers have been reported for human endometrial stem/progenitor cells; however, the lack of convenient markers in the mouse has made experimental investigation into endometrial regeneration difficult. We recently identified endometrial epithelial, endothelial, and immune cells, which express a reporter for the stem/progenitor marker, mouse telomerase reverse transcriptase (mTert). In this study, we investigate the expression pattern of a green fluorescent protein (GFP) reporter for mTert promoter activity (mTert-GFP) in endometrial regeneration following a menses-like event. mTert-GFP expression marks subepithelial populations of T cells and mature macrophages and may play a role in immune cell regulated repair. Clusters of mTert-GFP-positive epithelial cells were identified close to areas of reepithelialization and possibly highlight a role for mTert in the repair and regeneration of the endometrial epithelium.
Collapse
Affiliation(s)
- Fiona L Cousins
- 1 The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- 2 Department of Obstetrics and Gynecology, School of Clinical Sciences at Monash Health, Monash University Faculty of Medicine, Clayton, Australia
| | - Dorien F O
- 1 The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Yih Rue Ong
- 1 The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - David T Breault
- 3 Division of Endocrinology, Boston Children's Hospital, Harvard Stem Cell Institute, Boston, Massachusetts
| | - James A Deane
- 1 The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- 2 Department of Obstetrics and Gynecology, School of Clinical Sciences at Monash Health, Monash University Faculty of Medicine, Clayton, Australia
| | - Caroline E Gargett
- 1 The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- 2 Department of Obstetrics and Gynecology, School of Clinical Sciences at Monash Health, Monash University Faculty of Medicine, Clayton, Australia
| |
Collapse
|
19
|
Evans J, Infusini G, Mcgovern J, Cuttle L, Webb A, Nebl T, Milla L, Kimble R, Kempf M, Andrews CJ, Leavesley D, Salamonsen LA. Menstrual fluid factors facilitate tissue repair: identification and functional action in endometrial and skin repair. FASEB J 2018; 33:584-605. [DOI: 10.1096/fj.201800086r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jemma Evans
- The Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational MedicineMonash University Clayton Victoria Australia
| | | | - Jacqui Mcgovern
- Institute of Health and Biomedical Innovation Brisbane Queensland Australia
| | - Leila Cuttle
- Centre for Children's Burns and Trauma ResearchSchool of Biomedical SciencesInstitute of Health and Biomedical InnovationCentre for Children's Health ResearchQueensland University of Technology Brisbane Queensland Australia
| | - Andrew Webb
- Walter and Eliza Hall Institute Parkville Victoria Australia
| | - Thomas Nebl
- Walter and Eliza Hall Institute Parkville Victoria Australia
| | - Liz Milla
- Walter and Eliza Hall Institute Parkville Victoria Australia
| | - Roy Kimble
- Centre for Children's Burns and Trauma ResearchCentre for Children's Health ResearchThe University of Queensland South Brisbane Queensland Australia
| | - Margit Kempf
- Centre for Children's Burns and Trauma ResearchCentre for Children's Health ResearchThe University of Queensland South Brisbane Queensland Australia
| | - Christine J. Andrews
- Centre for Children's Burns and Trauma ResearchCentre for Children's Health ResearchThe University of Queensland South Brisbane Queensland Australia
| | - David Leavesley
- Institute of Health and Biomedical Innovation Brisbane Queensland Australia
- Skin Research Institute of Singapore Singapore
| | - Lois A. Salamonsen
- The Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational MedicineMonash University Clayton Victoria Australia
| |
Collapse
|
20
|
Armstrong GM, Maybin JA, Murray AA, Nicol M, Walker C, Saunders PTK, Rossi AG, Critchley HOD. Endometrial apoptosis and neutrophil infiltration during menstruation exhibits spatial and temporal dynamics that are recapitulated in a mouse model. Sci Rep 2017; 7:17416. [PMID: 29234102 PMCID: PMC5727295 DOI: 10.1038/s41598-017-17565-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/24/2017] [Indexed: 01/06/2023] Open
Abstract
Menstruation is characterised by synchronous shedding and restoration of tissue integrity. An in vivo model of menstruation is required to investigate mechanisms responsible for regulation of menstrual physiology and to investigate common pathologies such as heavy menstrual bleeding (HMB). We hypothesised that our mouse model of simulated menstruation would recapitulate the spatial and temporal changes in the inflammatory microenvironment of human menses. Three regulatory events were investigated: cell death (apoptosis), neutrophil influx and cytokine/chemokine expression. Well-characterised endometrial tissues from women were compared with uteri from a mouse model (tissue recovered 0, 4, 8, 24 and 48 h after removal of a progesterone-secreting pellet). Immunohistochemistry for cleaved caspase-3 (CC3) revealed significantly increased staining in human endometrium from late secretory and menstrual phases. In mice, CC3 was significantly increased at 8 and 24 h post-progesterone-withdrawal. Elastase+ human neutrophils were maximal during menstruation; Ly6G+ mouse neutrophils were maximal at 24 h. Human endometrial and mouse uterine cytokine/chemokine mRNA concentrations were significantly increased during menstrual phase and 24 h post-progesterone-withdrawal respectively. Data from dated human samples revealed time-dependent changes in endometrial apoptosis preceding neutrophil influx and cytokine/chemokine induction during active menstruation. These dynamic changes were recapitulated in the mouse model of menstruation, validating its use in menstrual research.
Collapse
Affiliation(s)
- Gregory M Armstrong
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Jacqueline A Maybin
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Alison A Murray
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Moira Nicol
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Catherine Walker
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Philippa T K Saunders
- MRC Centre for Inflammation Research (CIR), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Adriano G Rossi
- MRC Centre for Inflammation Research (CIR), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Hilary O D Critchley
- MRC Centre for Reproductive Health (CRH), University of Edinburgh, The Queen's Medical Research Institute (QMRI), 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
21
|
Evidence for a dynamic role for mononuclear phagocytes during endometrial repair and remodelling. Sci Rep 2016; 6:36748. [PMID: 27827431 PMCID: PMC5101509 DOI: 10.1038/srep36748] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022] Open
Abstract
In women, endometrial breakdown, which is experienced as menstruation, is characterised by high concentrations of inflammatory mediators and immune cells which account for ~40% of the stromal compartment during tissue shedding. These inflammatory cells are known to play a pivotal role in tissue breakdown but their contribution to the rapid scarless repair of endometrium remains poorly understood. In the current study we used a mouse model of menstruation to investigate dynamic changes in mononuclear phagocytes during endometrial repair and remodelling. Menstruation was simulated in MacGreen mice to allow visualisation of CSF1R+ mononuclear phagocytes. Immunohistochemistry revealed dynamic spatio-temporal changes in numbers and location of CSF1R-EGFP+ cells and Ly6G+ neutrophils. Flow cytometry confirmed a striking increase in numbers of GFP+ cells during repair (24 h): influxed cells were 66% F4/80+Gr-1+ and 30% F4/80−Gr-1+. Immunostaining identified distinct populations of putative ‘classical’ monocytes (GFP+F4/80−), monocyte-derived macrophages (GFP+F4/80+) and a stable population of putative tissue-resident macrophages (GFP-F4/80+) localised to areas of breakdown, repair and remodelling respectively. Collectively, these data provide the first compelling evidence to support a role for different populations of monocytes/macrophages in endometrial repair and provide the platform for future studies on the role of these cells in scarless healing.
Collapse
|
22
|
Evans J, Salamonsen LA, Winship A, Menkhorst E, Nie G, Gargett CE, Dimitriadis E. Fertile ground: human endometrial programming and lessons in health and disease. Nat Rev Endocrinol 2016; 12:654-667. [PMID: 27448058 DOI: 10.1038/nrendo.2016.116] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human endometrium is a highly dynamic tissue that is cyclically shed, repaired, regenerated and remodelled, primarily under the orchestration of oestrogen and progesterone, in preparation for embryo implantation. Humans are among the very few species that menstruate and that, consequently, are equipped with unique cellular and molecular mechanisms controlling these cyclic processes. Many reproductive pathologies are specific to menstruating species, and studies in animal models rarely translate to humans. Abnormal remodelling and regeneration of the human endometrium leads to a range of reproductive complications. Furthermore, the processes regulating endometrial remodelling and implantation, including those controlling hormonal impact, breakdown and repair, stem/progenitor cell activation, inflammation and cell invasion have broad applications to other fields. This Review presents current knowledge regarding the normal and abnormal function of the human endometrium. The development of biomarkers for prediction of uterine diseases and pregnancy disorders and future avenues of investigation to improve fertility and enhance endometrial function are also discussed.
Collapse
Affiliation(s)
- Jemma Evans
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
- Department of Physiology, Monash University, Clayton, 3800, Australia
| | - Lois A Salamonsen
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, 3800, Australia
| | - Amy Winship
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
| | - Ellen Menkhorst
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
| | - Guiying Nie
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, Australia
| | - Caroline E Gargett
- Department of Obstetrics and Gynaecology, Monash University, Clayton, 3800, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, 3168, Australia
| | - Eva Dimitriadis
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Australia
- Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, 3800, Australia
| |
Collapse
|
23
|
Paiva P, Lockhart MG, Girling JE, Olshansky M, Woodrow N, Marino JL, Hickey M, Rogers PAW. Identification of genes differentially expressed in menstrual breakdown and repair. Mol Hum Reprod 2016; 22:898-912. [PMID: 27609758 DOI: 10.1093/molehr/gaw060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/30/2016] [Accepted: 09/02/2016] [Indexed: 12/26/2022] Open
Abstract
STUDY QUESTION Does the changing molecular profile of the endometrium during menstruation correlate with the histological profile of menstruation. SUMMARY ANSWER We identified several genes not previously associated with menstruation; on Day 2 of menstruation (early-menstruation), processes related to inflammation are predominantly up-regulated and on Day 4 (late-menstruation), the endometrium is predominantly repairing and regenerating. WHAT IS KNOWN ALREADY Menstruation is induced by progesterone withdrawal at the end of the menstrual cycle and involves endometrial tissue breakdown, regeneration and repair. Perturbations in the regulation of menstruation may result in menstrual disorders including abnormal uterine bleeding. STUDY DESIGN, SIZE DURATION Endometrial samples were collected by Pipelle biopsy on Days 2 (n = 9), 3 (n = 9) or 4 (n = 6) of menstruation. PARTICIPANTS/MATERIALS, SETTING, METHODS RNA was extracted from endometrial biopsies and analysed by genome wide expression Illumina Sentrix Human HT12 arrays. Data were analysed using 'Remove Unwanted Variation-inverse (RUV-inv)'. Ingenuity pathway analysis (IPA) and the Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.7 were used to identify canonical pathways, upstream regulators and functional gene clusters enriched between Days 2, 3 and 4 of menstruation. Selected individual genes were validated by quantitative PCR. MAIN RESULTS AND THE ROLE OF CHANCE Overall, 1753 genes were differentially expressed in one or more comparisons. Significant canonical pathways, gene clusters and upstream regulators enriched during menstrual bleeding included those associated with immune cell trafficking, inflammation, cell cycle regulation, extracellular remodelling and the complement and coagulation cascade. We provide the first evidence for a role for glutathione-mediated detoxification (glutathione-S-transferase mu 1 and 2; GSTM1 and GSTM2) during menstruation. The largest number of differentially expressed genes was between Days 2 and 4 of menstruation (n = 1176). We identified several genes not previously associated with menstruation including lipopolysaccharide binding protein, serpin peptidase inhibitor, clade B (ovalbumin), member 3 (SERPINB3) and -4 (SERPINB4), interleukin-17C (IL17C), V-set domain containing T-cell activation inhibitor 1 (VTCN1), proliferating cell nuclear antigen factor (KIAA0101/PAF), trefoil factor 3 (TFF3), laminin alpha 2 (LAMA2) and serine peptidase inhibitor, Kazal type 1 (SPINK1). Genes related to inflammatory processes were up-regulated on Day 2 (early-menstruation), and those associated with endometrial repair and regeneration were up-regulated on Day 4 (late-menstruation). LIMITATIONS, REASONS FOR CAUTION Participants presented with a variety of endometrial pathologies related to bleeding status and other menstrual characteristics. These variations may also have influenced the menstrual process. WIDER IMPLICATIONS OF THE FINDINGS The temporal molecular profile of menstruation presented in this study identifies a number of genes not previously associated with the menstrual process. Our findings provide valuable insight into the menstrual process and may present novel targets for therapeutic intervention in cases of endometrial dysfunction. LARGE SCALE DATA All microarray data have been deposited in the public data repository Gene Expression Omnibus (GSE86003). STUDY FUNDING AND COMPETING INTERESTS Funding for this work was provided by a National Health and Medical Research Council of Australia (NHMRC) Project Grant APP1008553 to M.H., P.R. and J.G. M.H. is supported by an NHMRC Practitioner Fellowship. P.P. is supported by a NHMRC Early Career Fellowship. The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- Premila Paiva
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| | - Michelle G Lockhart
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| | - Jane E Girling
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| | - Moshe Olshansky
- Bioinformatics Division, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Microbiology, Monash University, Wellington Road and Blackburn Road, Clayton, VIC 3800, Australia
| | - Nicole Woodrow
- Pauline Gandel Imaging Centre, Royal Women's Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
| | - Jennifer L Marino
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| | - Martha Hickey
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| | - Peter A W Rogers
- Department of Obstetrics and Gynaecology, The University of Melbourne, Gynaecology Research Centre, Royal Women's Hospital, Cnr Flemington Rd and Grattan St, Parkville, VIC 3052, Australia
| |
Collapse
|
24
|
Cousins FL, Kirkwood PM, Murray AA, Collins F, Gibson DA, Saunders PTK. Androgens regulate scarless repair of the endometrial “wound” in a mouse model of menstruation. FASEB J 2016; 30:2802-11. [DOI: 10.1096/fj.201600078r] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/12/2016] [Indexed: 01/18/2023]
Affiliation(s)
- Fiona L. Cousins
- Medical Research Council (MRC) Centre for Inflammation ResearchThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Phoebe M. Kirkwood
- Medical Research Council (MRC) Centre for Inflammation ResearchThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Alison A. Murray
- MRC Centre for Reproductive HealthThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Frances Collins
- Medical Research Council (MRC) Centre for Inflammation ResearchThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Douglas A. Gibson
- Medical Research Council (MRC) Centre for Inflammation ResearchThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Philippa T. K. Saunders
- Medical Research Council (MRC) Centre for Inflammation ResearchThe University of Edinburgh, Queen's Medical Research InstituteEdinburghUnited Kingdom
| |
Collapse
|
25
|
Gargett CE, Schwab KE, Deane JA. Endometrial stem/progenitor cells: the first 10 years. Hum Reprod Update 2015; 22:137-63. [PMID: 26552890 PMCID: PMC4755439 DOI: 10.1093/humupd/dmv051] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The existence of stem/progenitor cells in the endometrium was postulated many years ago, but the first functional evidence was only published in 2004. The identification of rare epithelial and stromal populations of clonogenic cells in human endometrium has opened an active area of research on endometrial stem/progenitor cells in the subsequent 10 years. METHODS The published literature was searched using the PubMed database with the search terms ‘endometrial stem cells and menstrual blood stem cells' until December 2014. RESULTS Endometrial epithelial stem/progenitor cells have been identified as clonogenic cells in human and as label-retaining or CD44+ cells in mouse endometrium, but their characterization has been modest. In contrast, endometrial mesenchymal stem/stromal cells (MSCs) have been well characterized and show similar properties to bone marrow MSCs. Specific markers for their enrichment have been identified, CD146+PDGFRβ+ (platelet-derived growth factor receptor beta) and SUSD2+ (sushi domain containing-2), which detected their perivascular location and likely pericyte identity in endometrial basalis and functionalis vessels. Transcriptomics and secretomics of SUSD2+ cells confirm their perivascular phenotype. Stromal fibroblasts cultured from endometrial tissue or menstrual blood also have some MSC characteristics and demonstrate broad multilineage differentiation potential for mesodermal, endodermal and ectodermal lineages, indicating their plasticity. Side population (SP) cells are a mixed population, although predominantly vascular cells, which exhibit adult stem cell properties, including tissue reconstitution. There is some evidence that bone marrow cells contribute a small population of endometrial epithelial and stromal cells. The discovery of specific markers for endometrial stem/progenitor cells has enabled the examination of their role in endometrial proliferative disorders, including endometriosis, adenomyosis and Asherman's syndrome. Endometrial MSCs (eMSCs) and menstrual blood stromal fibroblasts are an attractive source of MSCs for regenerative medicine because of their relative ease of acquisition with minimal morbidity. Their homologous and non-homologous use as autologous and allogeneic cells for therapeutic purposes is currently being assessed in preclinical animal models of pelvic organ prolapse and phase I/II clinical trials for cardiac failure. eMSCs and stromal fibroblasts also exhibit non-stem cell-associated immunomodulatory and anti-inflammatory properties, further emphasizing their desirable properties for cell-based therapies. CONCLUSIONS Much has been learnt about endometrial stem/progenitor cells in the 10 years since their discovery, although several unresolved issues remain. These include rationalizing the terminology and diagnostic characteristics used for distinguishing perivascular stem/progenitor cells from stromal fibroblasts, which also have considerable differentiation potential. The hierarchical relationship between clonogenic epithelial progenitor cells, endometrial and decidual SP cells, CD146+PDGFR-β+ and SUSD2+ cells and menstrual blood stromal fibroblasts still needs to be resolved. Developing more genetic animal models for investigating the role of endometrial stem/progenitor cells in endometrial disorders is required, as well as elucidating which bone marrow cells contribute to endometrial tissue. Deep sequencing and epigenetic profiling of enriched populations of endometrial stem/progenitor cells and their differentiated progeny at the population and single-cell level will shed new light on the regulation and function of endometrial stem/progenitor cells.
Collapse
Affiliation(s)
- Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Road, Clayton 3168, Victoria, Australia
| | - Kjiana E Schwab
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Victoria, Australia
| | - James A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Road, Clayton 3168, Victoria, Australia
| |
Collapse
|
26
|
Maybin JA, Critchley HOD. Menstrual physiology: implications for endometrial pathology and beyond. Hum Reprod Update 2015; 21:748-61. [PMID: 26253932 PMCID: PMC4594618 DOI: 10.1093/humupd/dmv038] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/08/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Each month the endometrium becomes inflamed, and the luminal portion is shed during menstruation. The subsequent repair is remarkable, allowing implantation to occur if fertilization takes place. Aberrations in menstrual physiology can lead to common gynaecological conditions, such as heavy or prolonged bleeding. Increased knowledge of the processes involved in menstrual physiology may also have translational benefits at other tissue sites. METHODS Pubmed and Cochrane databases were searched for all original and review articles published in English until April 2015. Search terms included ‘endometrium’, ‘menstruation’, ‘endometrial repair’, ‘endometrial regeneration’ ‘angiogenesis’, ‘inflammation’ and ‘heavy menstrual bleeding’ or ‘menorrhagia’. RESULTS Menstruation occurs naturally in very few species. Human menstruation is thought to occur as a consequence of preimplantation decidualization, conferring embryo selectivity and the ability to adapt to optimize function. We highlight how current and future study of endometrial inflammation, vascular changes and repair/regeneration will allow us to identify new therapeutic targets for common gynaecological disorders. In addition, we describe how increased knowledge of this endometrial physiology will have many translational applications at other tissue sites. We highlight the clinical applications of what we know, the key questions that remain and the scientific and medical possibilities for the future. CONCLUSIONS The study of menstruation, in both normal and abnormal scenarios, is essential for the production of novel, acceptable medical treatments for common gynaecological complaints. Furthermore, collaboration and communication with specialists in other fields could significantly advance the therapeutic potential of this dynamic tissue.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Hilary O D Critchley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
27
|
Gómez E, Ruíz-Alonso M, Miravet J, Simón C. Human Endometrial Transcriptomics: Implications for Embryonic Implantation. Cold Spring Harb Perspect Med 2015; 5:a022996. [PMID: 25818663 PMCID: PMC4484960 DOI: 10.1101/cshperspect.a022996] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Human endometrium has been extensively investigated in the search for markers capable of predicting its receptive status. The completion of the Human Genome Project has triggered a rapid development of new fields in molecular biology, the "transcriptomics" being a major turning point in the knowledge acquisition of endometrial receptivity. Based on this, a customized Endometrial Receptivity Array (ERA) has been developed, which is capable of identifying the genomic signature of receptivity. This diagnostic tool showed that the window of implantation (WOI) is displaced in one out of four patients with implantation failure, allowing the identification of their personalized WOI. This strategy allows performing a personalized embryo transfer (pET) on the day in which the endometrium is receptive. The combination of a systems biology approach and next-generation sequencing will overcome the limitations of microarrays, and will, in the future, allow elucidation of the mechanisms involved in embryo implantation.
Collapse
Affiliation(s)
- Eva Gómez
- IGENOMIX, Parc Cientific Valencia University, Paterna, 46980 Valencia, Spain
| | - Maria Ruíz-Alonso
- IGENOMIX, Parc Cientific Valencia University, Paterna, 46980 Valencia, Spain
| | - Jose Miravet
- IGENOMIX, Parc Cientific Valencia University, Paterna, 46980 Valencia, Spain
| | - Carlos Simón
- IGENOMIX, Parc Cientific Valencia University, Paterna, 46980 Valencia, Spain Fundación Instituto Valenciano de Infertilidad (FIVI), Department of Obstetrics and Gynecology, School of Medicine, Valencia University and Instituto Universitario IVI/INCLIVA, 46010 Valencia, Spain Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
28
|
Coudyzer P, Lemoine P, Po C, Jordan BF, Van Der Smissen P, Courtoy PJ, Henriet P, Marbaix E. Induction of post-menstrual regeneration by ovarian steroid withdrawal in the functionalis of xenografted human endometrium. Hum Reprod 2015; 30:1156-68. [PMID: 25750204 DOI: 10.1093/humrep/dev043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION Does the endometrial functionalis have the potential to undergo self-renewal after menstruation and how is this process controlled by ovarian steroids? SUMMARY ANSWER Endometrial xenografts subjected to withdrawal of estradiol and progesterone shrink but also show signs of proliferation and tissue repair; new estradiol supply prevents atrophy but is not sufficient to increase graft volume. WHAT IS KNOWN ALREADY Menstruation, i.e. cyclic proteolysis of the extracellular matrix of endometrial functionalis, is induced by a fall in estrogen and progesterone concentration and is followed by tissue regeneration. However, there is debate about whether regenerating cells must originate from the basalis or from stem cells and whether new estrogen supply is required for the early repair concomitant with menstruation. STUDY DESIGN, SIZE, DURATION Fragments from human endometrial functionalis (from 24 hysterectomy specimens) were xenografted in ovariectomized SCID mice and submitted to a 4-day estradiol and progesterone withdrawal (to mimic menstruation) followed by re-exposure to estradiol (to mimic the proliferative phase). We measured signs of proliferation and changes in graft volume. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrium was collected from spontaneously cycling women. Cell proliferation was examined by immunolabeling Ki-67, cyclin D1 and phosphorylated-histone H3. Xenograft volume was measured by magnetic resonance imaging. Xenograft histomorphometry was performed to determine how the different tissue compartments contributed to volume change. MAIN RESULTS AND THE ROLE OF CHANCE Hormone withdrawal induced a rapid decrease in graft volume mainly attributable to stroma condensation and breakdown, concomitant with an increase of proliferation markers. Reinsertion of estradiol pellets after induced menstruation blocked volume decrease and stimulated epithelial and stromal growth, but, surprisingly, did not induce graft enlargement. Reinsertion of both estradiol and progesterone pellets blocked apoptosis. LIMITATIONS, REASONS FOR CAUTION Mechanisms of endometrial remodeling are different in women and mice and the contribution of circulating inflammatory cells in both species remains to be clarified. Moreover, during human menstruation, endometrial fragments resulting from tissue proteolysis can be expelled by the menstrual flow, unlike in this model. WIDER IMPLICATIONS OF THE FINDINGS Menstruation is a multifocal event within the functionalis. This is the first evidence that endometrial fragments that are not shed after menstrual tissue breakdown can support endometrial regeneration. Endometriosis is commonly thought to result from the retrograde migration of menstrual fragments of the degraded functionalis into the peritoneal cavity. Our study supports their potential to regenerate as ectopic endometrium. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Fonds de la Recherche Scientifique Médicale, Concerted Research Actions, Communauté Française de Belgique, Région wallonne, Région bruxelloise and Loterie nationale. P.H. and B.F.J. are research associates of the Belgian Fonds de la Recherche Scientifique (F.R.S.-F.N.R.S.). E.M. is Associate Editor at Human Reproduction. There is no conflict of interest to declare.
Collapse
Affiliation(s)
- Pauline Coudyzer
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Pascale Lemoine
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Chrystelle Po
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Etienne Marbaix
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
29
|
Sheldon RE, Shmygol A, Van Den Berg HA, Blanks AM. Functional and morphological development of the womb throughout life. Sci Prog 2015; 98:103-27. [PMID: 26288915 PMCID: PMC10365438 DOI: 10.3184/003685015x14308363103415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The uterus undergoes changes throughout a woman's life, beginning with her own embryonic development when she is still in the womb, commencing a monthly cycle at the onset of adulthood, and undergoing dramatic changes during pregnancy and parturition. The impact of preterm labour and other perinatal health problems is significant, both in human and financial terms; therefore the study of the physiological and regulatory changes which the uterus undergoes can be of enormous potential benefit. Here we briefly review the current state of knowledge, with an emphasis on the importance of changes in connectivity in the uterine smooth muscle cell network and on recent mathematical modelling work aimed at elucidating the role of spatial heterogeneity in this connected network.
Collapse
|
30
|
Affiliation(s)
- Tetsuo Maruyama
- Department of Obstetrics and Gynecology; School of Medicine; Keio University; Tokyo Japan
| |
Collapse
|
31
|
Zhu H, Hou CC, Luo LF, Hu YJ, Yang WX. Endometrial stromal cells and decidualized stromal cells: origins, transformation and functions. Gene 2014; 551:1-14. [PMID: 25168894 DOI: 10.1016/j.gene.2014.08.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 07/24/2014] [Accepted: 08/24/2014] [Indexed: 10/24/2022]
Abstract
Decidualization of endometrium, which is characterized by endometrial stromal cell (ESC) decidualization, vascular reconstruction, immune cell recruitment, and plentiful molecule production, is a crucial step for uterus to become receptive for embryo. When implantation takes place, ESCs surround and directly interact with embryo. Decidualized stromal cells (DSCs) are of great importance in endometrial decidualization, having a broad function in regulating immune activity and vascular remodeling of uterus. DSCs are shown to have a higher metabolic level and looser cytoskeleton than ESCs. What's the origin of ESCs and how ESCs successfully transform into DSCs had puzzled scientists in the last decades. Breakthrough had been achieved recently, and many studies had elucidated some of the characters and functions of DSCs. However, several questions still remain unclear. This paper reviews current understanding of where ESCs come from and how ESCs differentiate into DSCs, summarizes some characters and functions of DSCs, analyzes current studies and their limitations and points out research areas that need further investigation.
Collapse
Affiliation(s)
- Ha Zhu
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong-Cong Hou
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ling-Feng Luo
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan-Jun Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China.
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
32
|
Cominelli A, Gaide Chevronnay HP, Lemoine P, Courtoy PJ, Marbaix E, Henriet P. Matrix metalloproteinase-27 is expressed in CD163+/CD206+ M2 macrophages in the cycling human endometrium and in superficial endometriotic lesions. Mol Hum Reprod 2014; 20:767-75. [PMID: 24810263 DOI: 10.1093/molehr/gau034] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix remodelling. In the human endometrium, the expression and activity of several MMPs are maximal during the menstrual phase. Moreover, MMPs are thought to be involved in the pathogenesis of endometriosis and cancers, in particular with invasion and metastasis. We recently reported that MMP-27 is a unique MMP with an intracellular retention motif. We investigated the expression and cellular localization of MMP-27 in the cycling human endometrium and in endometriotic lesions. MMP-27 mRNA was detected throughout the menstrual cycle. Despite large interpatient variations, mRNA levels increased from the proliferative to the secretory phase, to peak during the menstrual phase. MMP-27 was immunolocalized in large isolated cells scattered throughout the stroma and around blood vessels: these cells were most abundant at menstruation and were identified by immunofluorescence as CD45(+), CD163(+) and CD206(+) macrophages. CD163(+) macrophages were also abundant in endometriotic lesions, but showed different patterns in ovarian or peritoneal endometriotic lesions (co-labelling for CD206 and MMP-27) and rectovaginal lesions (no co-labelling). In conclusion, MMP-27 is expressed in a subset of endometrial macrophages related to menstruation and in ovarian and peritoneal endometriotic lesions.
Collapse
Affiliation(s)
- Antoine Cominelli
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | - Pascale Lemoine
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Etienne Marbaix
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
33
|
Cominelli A, Halbout M, N'Kuli F, Lemoine P, Courtoy PJ, Marbaix E, Tyteca D, Henriet P. A unique C-terminal domain allows retention of matrix metalloproteinase-27 in the endoplasmic reticulum. Traffic 2014; 15:401-17. [PMID: 24548619 DOI: 10.1111/tra.12149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 11/29/2022]
Abstract
Matrix metalloproteinase-27 (MMP-27) is poorly characterized. Sequence comparison suggests that a C-terminal extension (CTE) includes a potential transmembrane domain as in some membrane-type (MT)-MMPs. Having noticed that MMP-27 was barely secreted, we investigated its subcellular localization and addressed CTE contribution for MMP-27 retention. Intracellular MMP-27 was sensitive to endoglycosidase H. Subcellular fractionation and confocal microscopy evidenced retention of endogenous MMP-27 or recombinant rMMP-27 in the endoplasmic reticulum (ER) with locked exit across the intermediate compartment (ERGIC). Conversely, truncated rMMP-27 without CTE accessed downstream secretory compartments (ERGIC and Golgi) and was constitutively secreted. CTE addition to rMMP-10 (a secreted MMP) caused ER retention and blocked secretion. Addition of a PKA target sequence to the cytosolic C-terminus of transmembrane MT1-MMP/MMP-14 led to effective phosphorylation upon forskolin stimulation, but not for MMP-27, excluding transmembrane anchorage. Moreover, MMP-27 was protected from digestion by proteinase K. Finally, MT1-MMP/MMP-14 but neither endogenous nor recombinant MMP-27 partitioned in the detergent phase after Triton X-114 extraction, indicating that MMP-27 is not an integral membrane protein. In conclusion, MMP-27 is efficiently retained within the ER due to its unique CTE, which does not lead to stable membrane insertion. This could represent a novel ER retention system.
Collapse
Affiliation(s)
- Antoine Cominelli
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, Box B1.75.05, B-1200, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Cousins FL, Murray A, Esnal A, Gibson DA, Critchley HOD, Saunders PTK. Evidence from a mouse model that epithelial cell migration and mesenchymal-epithelial transition contribute to rapid restoration of uterine tissue integrity during menstruation. PLoS One 2014; 9:e86378. [PMID: 24466063 PMCID: PMC3899239 DOI: 10.1371/journal.pone.0086378] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/10/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In women dynamic changes in uterine tissue architecture occur during each menstrual cycle. Menses, characterised by the shedding of the upper functional layer of the endometrium, is the culmination of a cascade of irreversible changes in tissue function including stromal decidualisation, inflammation and production of degradative enzymes. The molecular mechanisms that contribute to the rapid restoration of tissue homeostasis at time of menses are poorly understood. METHODOLOGY A modified mouse model of menses was developed to focus on the events occurring within the uterine lining during endometrial shedding/repair. Decidualisation, vaginal bleeding, tissue architecture and cell proliferation were evaluated at 4, 8, 12, and 24 hours after progesterone (P4) withdrawal; mice received a single injection of bromodeoxyuridine (BrdU) 90 mins before culling. Expression of genes implicated in the regulation of mesenchymal to epithelial transition (MET) was determined using a RT2 PCR profiler array, qRTPCR and bioinformatic analysis. PRINCIPAL FINDINGS Mice exhibited vaginal bleeding between 4 and 12 hours after P4 withdrawal, concomitant with detachment of the decidualised cell mass from the basal portion of the endometrial lining. Immunostaining for BrdU and pan cytokeratin revealed evidence of epithelial cell proliferation and migration. Cells that appeared to be in transition from a mesenchymal to an epithelial cell identity were identified within the stromal compartment. Analysis of mRNAs encoding genes expressed exclusively in the epithelial or stromal compartments, or implicated in MET, revealed dynamic changes in expression, consistent with a role for reprogramming of mesenchymal cells so that they could contribute to re-epithelialisation. CONCLUSIONS/SIGNIFICANCE These studies have provided novel insights into the cellular processes that contribute to re-epithelialisation post-menses implicating both epithelial cell migration and mesenchymal cell differentiation in restoration of an intact epithelial cell layer. These insights may inform development of new therapies to induce rapid healing in the endometrium and other tissues and offer hope to women who suffer from heavy menstrual bleeding.
Collapse
Affiliation(s)
- Fiona L. Cousins
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Alison Murray
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Arantza Esnal
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Douglas A. Gibson
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Hilary O. D. Critchley
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Philippa T. K. Saunders
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Coudyzer P, Lemoine P, Jordan BF, Gallez B, Galant C, Nisolle M, Courtoy PJ, Henriet P, Marbaix E. Hypoxia is not required for human endometrial breakdown or repair in a xenograft model of menstruation. FASEB J 2013; 27:3711-9. [DOI: 10.1096/fj.13-232074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Pauline Coudyzer
- Cell Biology Unitde Duve InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Pascale Lemoine
- Cell Biology Unitde Duve InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Bénédicte F. Jordan
- Biomedical Magnetic Resonance Research GroupLouvain Drug Research InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research GroupLouvain Drug Research InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Christine Galant
- Pathology DepartmentInstitut de Recherche Expérimentale et CliniqueUniversité Catholique de LouvainBrusselsBelgium
| | - Michelle Nisolle
- Laboratory of Tumor and Development BiologyGroupe Interdisciplinaire de Génoprotéomique AppliquéeUniversité de LiègeLiègeBelgium
| | - Pierre J. Courtoy
- Cell Biology Unitde Duve InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Patrick Henriet
- Cell Biology Unitde Duve InstituteUniversité Catholique de LouvainBrusselsBelgium
| | - Etienne Marbaix
- Cell Biology Unitde Duve InstituteUniversité Catholique de LouvainBrusselsBelgium
| |
Collapse
|
36
|
Abstract
The ovarian steroid hormones progesterone and estradiol are well established regulators of human endometrial function. However, more recent evidence suggests that androgens and locally generated steroids, such as the glucocorticoids, also have a significant impact on endometrial breakdown and repair. The temporal and spatial pattern of steroid receptor presence in endometrial cells has a significant impact on the endometrial response to steroids. Furthermore, regulation of steroid receptor function by modulatory proteins further refines local responses. This review focuses on steroid regulation of endometrial function during the luteo-follicular transition with a focus on menstruation and endometrial repair.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | | |
Collapse
|
37
|
Abstract
The functional layer of the human endometrium is a highly regenerative tissue undergoing monthly cycles of growth, differentiation and shedding during a woman's reproductive years. Fluctuating levels of circulating estrogen and progesterone orchestrate this dramatic remodeling of human endometrium. The thin inactive endometrium of postmenopausal women which resembles the permanent basal layer of cycling endometrium retains the capacity to respond to exogenous sex steroid hormones to regenerate into a thick functional endometrium capable of supporting pregnancy. Endometrial regeneration also follows parturition and endometrial resection. In non menstruating rodents, endometrial epithelium undergoes rounds of proliferation and apoptosis during estrus cycles. The recent identification of adult stem cells in both human and mouse endometrium suggests that epithelial progenitor cells and the mesenchymal stem/stromal cells have key roles in the cyclical regeneration of endometrial epithelium and stroma. This review will summarize the evidence for endometrial stem/progenitor cells, examine their role in mouse models of endometrial epithelial repair and estrogen-induced endometrial regeneration, and also describe the generation of endometrial-like epithelium from human embryonic stem cells. With markers now available for identifying endometrial mesenchymal stem/stromal cells, their possible role in gynecological diseases associated with abnormal endometrial proliferation and their potential application in cell-based therapies to regenerate reproductive and other tissues will be discussed.
Collapse
Affiliation(s)
- Caroline E Gargett
- The Ritchie Centre, Monash Institute of Medical Research, Clayton, Victoria, Australia.
| | | | | |
Collapse
|
38
|
Henriet P, Gaide Chevronnay HP, Marbaix E. The endocrine and paracrine control of menstruation. Mol Cell Endocrinol 2012; 358:197-207. [PMID: 21820486 DOI: 10.1016/j.mce.2011.07.042] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 01/29/2023]
Abstract
During the reproductive life, the human endometrium undergoes cycles of substantial remodeling including, at menstruation, a massive but delimited tissue breakdown immediately followed by scarless repair. The present review aims at summarizing the current knowledge on the endocrine and paracrine control of menstruation in the light of recent observations that undermine obsolete dogmas. Menstruation can be globally considered as a response to falling progesterone concentration. However, tissue breakdown is heterogeneous and tightly controlled in space and time by a complex network of regulators and effectors, including cytokines, chemokines, proteases and various components of an inflammatory response. Moreover, menstruation must be regarded as part of a complex and integrated mechanism of tissue remodeling including features that precede and follow tissue lysis, i.e. decidualization and immediate post-menstrual regeneration. The understanding of the regulation of menstruation is of major basic and clinical interest. Indeed, these mechanisms largely overlap with those controlling other histopathological occurrences of tissue remodeling, such as development and cancer, and inappropriate control of menstrual features is a major potential cause of two frequent endometrial pathologies (i.e. abnormal uterine bleeding and endometriosis).
Collapse
Affiliation(s)
- Patrick Henriet
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, avenue Hippocrate, 75, B-1200 Bruxelles, Belgium.
| | | | | |
Collapse
|
39
|
Al-Sabbagh M, Lam EWF, Brosens JJ. Mechanisms of endometrial progesterone resistance. Mol Cell Endocrinol 2012; 358:208-15. [PMID: 22085558 DOI: 10.1016/j.mce.2011.10.035] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 10/31/2011] [Accepted: 10/31/2011] [Indexed: 10/15/2022]
Abstract
Throughout the reproductive years, the rise and fall in ovarian hormones elicit in the endometrium waves of cell proliferation, differentiation, recruitment of inflammatory cells, apoptosis, tissue breakdown and regeneration. The activated progesterone receptor, a member of the superfamily of ligand-dependent transcription factors, is the master regulator of this intense tissue remodelling process in the uterus. Its activity is tightly regulated by interaction with cell-specific transcription factors and coregulators as well as by specific posttranslational modifications that respond dynamically to a variety of environmental and inflammatory signals. Endometriosis, a chronic inflammatory disorder, disrupts coordinated progesterone responses throughout the reproductive tract, including in the endometrium. This phenomenon is increasingly referred to as 'progesterone resistance'. Emerging evidence suggests that progesterone resistance in endometriosis is not just a consequence of perturbed progesterone signal transduction caused by chronic inflammation but associated with epigenetic chromatin changes that determine the intrinsic responsiveness of endometrial cells to differentiation cues.
Collapse
Affiliation(s)
- Marwa Al-Sabbagh
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | |
Collapse
|
40
|
Braundmeier AG, Dayger CA, Mehrotra P, Belton RJ, Nowak RA. EMMPRIN is secreted by human uterine epithelial cells in microvesicles and stimulates metalloproteinase production by human uterine fibroblast cells. Reprod Sci 2012; 19:1292-301. [PMID: 22729071 DOI: 10.1177/1933719112450332] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Endometrial remodeling is a physiological process involved in the gynecological disease, endometriosis. Tissue remodeling is directed by uterine fibroblast production of matrix metalloproteinases (MMPs). Several MMPs are regulated directly by the protein extracellular matrix metalloproteinase inducer (EMMPRIN) and also by proinflammatory cytokines such as interleukin (IL)1-α/β. We hypothesized that human uterine epithelial cells (HESs) secrete intact EMMPRIN to stimulate MMPs. Microvesicles from HES cell-conditioned medium (CM) expressed intact EMMPRIN protein. Treatment of HES cells with estradiol or phorbyl 12-myristate-13-acetate increased the release of EMMPRIN-containing microvesicles. The HES CM stimulated MMP-1, -2, and -3 messenger RNA levels in human uterine fibroblasts (HUFs) and EMMPRIN immunodepletion from HES-cell concentrated CM reduced MMP stimulation (P < .05). Treatment of HUF cells with low concentrations of IL-1β/α stimulated MMP production (P < .05). These results indicate that HES cells regulate MMP production by HUF cells by secretion of EMMPRIN, in response to ovarian hormones, proinflammatory cytokines as well as activation of protein kinase C.
Collapse
Affiliation(s)
- A G Braundmeier
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA.
| | | | | | | | | |
Collapse
|
41
|
Nguyen HPT, Sprung CN, Gargett CE. Differential expression of Wnt signaling molecules between pre- and postmenopausal endometrial epithelial cells suggests a population of putative epithelial stem/progenitor cells reside in the basalis layer. Endocrinology 2012; 153:2870-83. [PMID: 22474188 PMCID: PMC3359601 DOI: 10.1210/en.2011-1839] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 03/07/2012] [Indexed: 11/19/2022]
Abstract
The human endometrium undergoes extensive monthly regeneration in response to fluctuating levels of circulating estrogen and progesterone in premenopausal (Pre-M) women. In contrast, postmenopausal (Post-M) endometrium is thin and quiescent with low mitotic activity, similar to the Pre-M endometrial basalis layer. Clonogenic epithelial stem/progenitor (ESP) cells, likely responsible for regenerating endometrial epithelium, have been identified in Pre-M and Post-M endometrium, but their location is unknown. We undertook transcriptional profiling of highly purified epithelial cells from full-thickness Pre-M and Post-M endometrium to identify differentially regulated genes that may indicate a putative ESP cell population resides in the basalis of Pre-M and basalis-like Post-M endometrium. Of 1077 differentially expressed genes identified, the Wnt signaling pathway, important in endometrial development and stem cell regulation, was one of the main gene families detected, including 22 Wnt-associated genes. Twelve genes were validated using quantitative RT-PCR, and all were concordant with microarray data. Immunostaining showed glandular epithelial location of Wnt-regulated genes, Axin-related protein 2 and β-catenin. Axin2 localized to the nucleus of basalis Pre-M and Post-M and cytoplasm of functionalis Pre-M endometrium, suggesting that it regulates β-catenin. Comparison of our Post-M gene profile with published gene microarray datasets revealed similarities to Pre-M basalis epithelial profiles. This differential expression of multiple Wnt-associated genes in human Pre-M and Post-M endometrial epithelial cells and the similar gene profile of Post-M and Pre-M basalis epithelium suggests that a population of putative endometrial ESP may reside in the basalis of Pre-M endometrium, which may be responsible for regenerating glandular epithelium each month.
Collapse
Affiliation(s)
- Hong P. T. Nguyen
- The Ritchie Centre (H.P.T.N., C.E.G.) and Centre for Innate Immunity and Infectious Disease (C.N.S.), Monash Institute of Medical Research, and Department of Obstetrics and Gynaecology (H.P.T.N., C.E.G.), Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | | | | |
Collapse
|
42
|
Gargett CE, Ye L. Endometrial reconstruction from stem cells. Fertil Steril 2012; 98:11-20. [PMID: 22657248 DOI: 10.1016/j.fertnstert.2012.05.004] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/02/2012] [Accepted: 05/03/2012] [Indexed: 01/13/2023]
Abstract
Adult stem cells have been identified in the highly regenerative human endometrium on the basis of their functional attributes. They can reconstruct endometrial tissue in vivo suggesting their possible use in treating disorders associated with inadequate endometrium. The identification of specific markers for endometrial mesenchymal stem cells and candidate markers for epithelial progenitor cells enables the potential use of endometrial stem/progenitor cells in reconstructing endometrial tissue in Asherman syndrome and intrauterine adhesions.
Collapse
Affiliation(s)
- Caroline E Gargett
- The Ritchie Centre, Monash Institute of Medical Research, Clayton, Victoria, Australia.
| | | |
Collapse
|
43
|
Ruiz-Alonso M, Blesa D, Simón C. The genomics of the human endometrium. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1931-42. [PMID: 22634130 DOI: 10.1016/j.bbadis.2012.05.004] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 04/04/2012] [Accepted: 05/06/2012] [Indexed: 12/11/2022]
Abstract
The endometrium is a complex tissue that lines the inside of the endometrial cavity. The gene expression of the different endometrial cell types is regulated by ovarian steroids and paracrine-secreted molecules from neighbouring cells. Due to this regulation, the endometrium goes through cyclic modifications which can be divided simply into the proliferative phase, the secretory phase and the menstrual phase. Successful embryo implantation depends on three factors: embryo quality, the endometrium's state of receptivity, and a synchronised dialogue between the maternal tissue and the blastocyst. There is a need to characterise the endometrium's state of receptivity in order to prevent reproductive failure. No single molecular or histological marker for this status has yet been found. Here, we review the global transcriptomic analyses performed in the last decade on a normal human endometrium. These studies provide us with a clue about what global gene expression can be expected for a non-pathological endometrium. These studies have shown endometrial phase-specific transcriptomic profiles and common temporal gene expression patterns. We summarise the biological processes and genes regulated in the different phases of natural cycles and present other works on different conditions as well as a receptivity diagnostic tool based on a specific gene set profile. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.
Collapse
Affiliation(s)
- Maria Ruiz-Alonso
- Parc Cientific Universitat de Valencia, Calle Catedrático Agustín Escardino, Valencia, Spain.
| | | | | |
Collapse
|
44
|
Li YF, Xu XB, Chen XH, Wei G, He B, Wang JD. The nuclear factor-κB pathway is involved in matrix metalloproteinase-9 expression in RU486-induced endometrium breakdown in mice. Hum Reprod 2012; 27:2096-106. [PMID: 22587999 DOI: 10.1093/humrep/des110] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Progesterone-withdrawal (WP)-induced endometrial breakdown occurs in both physiological and pathological processes such as menstruation and abortion. However, the underlying mechanisms are not clearly understood. As the nuclear factor-κB (NF-κB) pathway has been proposed to play a role in endometrial breakdown, we tested this hypothesis using RU486-induced mouse menstruation-like model. METHODS The activation of NF-κB was evaluated by immunohistochemistry, western blot and immunofluorescence. The expression of matrix metalloproteinase-9 (MMP9) was analyzed by real-time PCR and its proteins by gelatin zymography and western blot. Chromatin immunoprecipitation was used to investigate the direct binding of NF-κB to MMP9 gene promoter. Inhibitors of NF-κB were used to block its signal in vivo and in vitro to analyze the function of NF-κB in the tissue breakdown process. RESULTS Administration of RU486 resulted in increased phospho-IκB levels and nuclear translocation of p65 in decidual stromal cells, accompanied by the up-regulation of NF-κB inducing kinase and IκB kinase β mRNA. The NF-κB inhibitor, 'pyrrolidine dithiocarbamate' partially suppressed the RU486-induced endometrial breakdown, thus verifying the role of this pathway in vivo. MMP9 was up- and down-regulated following the NF-κB activation and inhibition, respectively. RU486 stimulated recruitment of NF-κB p65 to the MMP9 promoter and further increased its expression. Effects of NF-κB activation and inactivation on MMP9 expression were further explored in human stromal cells in vitro. A similar MMP9 expression pattern was observed in cultured human, as well as mouse, decidual stromal cells following RU486 treatment. CONCLUSIONS The activation of the NF-κB pathway induces downstream target genes, including MMP9 from stromal cells to facilitate tissue breakdown in mouse uterus, highlighting the likelihood that this regulatory pattern exists in the human endometrium.
Collapse
Affiliation(s)
- Yun-Feng Li
- Graduate School of Peking Union Medical College, Beijing 100730, People's Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
Maybin J, Barcroft J, Thiruchelvam U, Hirani N, Jabbour H, Critchley H. The presence and regulation of connective tissue growth factor in the human endometrium. Hum Reprod 2012; 27:1112-21. [PMID: 22328559 PMCID: PMC3303491 DOI: 10.1093/humrep/der476] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 12/05/2011] [Accepted: 12/21/2011] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The human endometrium efficiently repairs each month after menstruation. The mechanisms involved in this repair process remain undefined. Aberrations in endometrial repair may lead to the common disorder of heavy menstrual bleeding. We hypothesized that connective tissue growth factor (CTGF) is increased at the time of endometrial repair post-menses and that this increase is regulated by prostaglandins (PGs) and hypoxic conditions present during menstruation. METHODS AND RESULTS Examination of 41 endometrial biopsies from 5 stages of the menstrual cycle revealed maximal CTGF mRNA expression (using quantitative RT-PCR) at menstruation and peak protein levels during the proliferative phase. CTGF was immunolocalized to epithelial and stromal cells, with intense staining of occasional stromal cells during the proliferative phase. Dual immunohistochemistry identified these cells as macrophages. Treatment of endometrial epithelial cells with 100 nM PGE(2), PGF(2α) or hypoxia (0.5% O(2)) revealed a significant increase in CTGF mRNA expression (P < 0.01 for all, versus vehicle control). Cells treated simultaneously with PGE(2) and hypoxia revealed a synergistic increase in CTGF expression (P < 0.05 versus PGE(2) or hypoxia alone) and maximal secreted CTGF protein levels (P < 0.05 versus control). CONCLUSIONS CTGF is increased in the human endometrium at the time of endometrial repair post-menses. The increase in CTGF may be mediated by PG production and the transient hypoxic episode observed in the endometrium at menstruation.
Collapse
Affiliation(s)
| | | | | | | | | | - H.O.D. Critchley
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
46
|
Na KH, Lee HJ, Choi JH, Eun JW, Nam SW, Yoon TK, Kim GJ. Dynamic alterations in integrin α4 expression by hypoxia are involved in trophoblast invasion during early implantation. J Cell Biochem 2012; 113:685-94. [DOI: 10.1002/jcb.23398] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Fauser BCJM, Laven JSE, Tarlatzis BC, Moley KH, Critchley HOD, Taylor RN, Berga SL, Mermelstein PG, Devroey P, Gianaroli L, D'Hooghe T, Vercellini P, Hummelshoj L, Rubin S, Goverde AJ, De Leo V, Petraglia F. Sex steroid hormones and reproductive disorders: impact on women's health. Reprod Sci 2011; 18:702-12. [PMID: 21795737 DOI: 10.1177/1933719111405068] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of sex steroid hormones in reproductive function in women is well established. However, in the last two decades it has been shown that receptors for estrogens, progesterone and androgens are expressed in non reproductive tissue /organs (bone, brain, cardiovascular system) playing a role in their function. Therefore, it is critical to evaluate the impact of sex steroid hormones in the pathophysiology of some diseases (osteoporosis, Alzheimer, atherosclerosis). In particular, women with primary ovarian insufficiency, polycystic ovary syndrome, endometriosis and climacteric syndrome may have more health problems and therefore an hormonal treatment may be crucial for these women.
Collapse
Affiliation(s)
- Bart C J M Fauser
- 1Department of Reproductive Medicine and Gynecology, University Medical Center, Utrecht, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Maybin JA, Battersby S, Hirani N, Nikitenko LL, Critchley HOD, Jabbour HN. The expression and regulation of adrenomedullin in the human endometrium: a candidate for endometrial repair. Endocrinology 2011; 152:2845-56. [PMID: 21558311 PMCID: PMC3192419 DOI: 10.1210/en.2010-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 04/21/2011] [Indexed: 12/29/2022]
Abstract
After menstruation, the endometrium has a remarkable capacity for repair, but the factors involved remain undefined. We hypothesize adrenomedullin (AM) plays a role in this process. Premenstrually progesterone levels decline, stimulating prostaglandin (PG) synthesis, vasoconstriction, and hypoxia. This study aimed to determine 1) AM expression throughout the menstrual (M) cycle and 2) its regulation by PG and hypoxia. Human endometrial biopsies (n = 51) were collected with ethical approval and consent. AM mRNA expression was examined by quantitative RT-PCR and was found to be selectively elevated in endometrium from the menstrual (M) phase (P < 0.001). AM immunohistochemical staining was maximal in M and proliferative (P) endometrium. Culture of secretory, but not P, explants with 100 nm PGF(2α) or hypoxia (0.5% O2) increased AM mRNA (P < 0.05). P explants were induced to increase AM expression using in vitro progesterone withdrawal but required the presence of hypoxia (P < 0.05). Short hairpin sequences against hypoxia-inducible factor-1α (HIF-1α) inhibited AM hypoxic up-regulation but did not alter PGF(2α)-induced expression. The AM receptor was immunolocalized to endothelial cells in both lymphatic and blood vessels. Conditioned medium from PGF(2α)-treated cells increased endothelial cell proliferation and branching (P < 0.05). This was abolished by AM receptor antagonists. In conclusion, AM is elevated at the time of endometrial repair and induces both angiogenesis and lymphangiogenesis by stimulating endothelial cell proliferation and tube formation. In the human endometrium, AM expression is up-regulated by two mechanisms: a HIF-1α-mediated hypoxic induction and a HIF-1α-independent PGF(2α) pathway. These physiological mechanisms may provide novel therapeutic targets for disorders such as heavy menstrual bleeding.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- Centre for Reproductive Biology, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
49
|
Maybin JA, Hirani N, Jabbour HN, Critchley HO. Novel roles for hypoxia and prostaglandin E2 in the regulation of IL-8 during endometrial repair. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1245-56. [PMID: 21356375 PMCID: PMC3047791 DOI: 10.1016/j.ajpath.2010.11.070] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 10/20/2010] [Accepted: 11/29/2010] [Indexed: 11/15/2022]
Abstract
The endometrium has a remarkable capacity for efficient repair; however, factors involved remain undefined. Premenstrual progesterone withdrawal leads to increased prostaglandin (PG) production and local hypoxia. Here we determined human endometrial expression of interleukin-8 (IL-8) and the roles of PGE(2) and hypoxia in its regulation. Endometrial biopsy specimens (n = 51) were collected. Endometrial cells and explants were exposed to 100 nmol/L of PGE(2) or 0.5% O(2). The endometrial IL-8 concentration peaked during menstruation (P < 0.001) and had a significant proangiogenic effect. IL-8 was increased by PGE(2) and hypoxia in secretory but not proliferative explants, which suggests that exposure to progesterone is essential. In vitro progesterone withdrawal induced significant IL-8 up-regulation in proliferative explants primed with progestins, but only in the presence of hypoxia. Epithelial cells treated simultaneously with PGE(2) and hypoxia demonstrated synergistic increases in IL-8. Inhibition of HIF-1 by short hairpin RNA abolished hypoxic IL-8 induction, and inhibition of NF-κB by an adenoviral dominant negative inhibitor decreased PGE(2)-induced IL-8 expression (P > 0.05). Increased menstrual IL-8 is consistent with a role in repair. Progesterone withdrawal, hypoxia, and PGE(2) regulate endometrial IL-8 by acting via HIF-1 and NF-κB. Hence, progesterone withdrawal may activate two distinct pathways to initiate endometrial repair.
Collapse
Affiliation(s)
- Jacqueline A. Maybin
- University of Edinburgh Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, Scotland
| | - Nikhil Hirani
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh, Scotland
| | - Henry N. Jabbour
- MRC Human Reproductive Sciences Unit, The Queen's Medical Research Institute, Edinburgh, Scotland
| | - Hilary O.D. Critchley
- University of Edinburgh Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, Scotland
| |
Collapse
|
50
|
Affiliation(s)
- Lois A Salamonsen
- Prince Henry's Institute of Medical Research, Melbourne, Victoria 3168, Australia.
| | | |
Collapse
|