10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      LEFTY2 inhibits endometrial receptivity by downregulating Orai1 expression and store-operated Ca 2+ entry

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Abstract

          Early embryo development and endometrial differentiation are initially independent processes, and synchronization, imposed by a limited window of implantation, is critical for reproductive success. A putative negative regulator of endometrial receptivity is LEFTY2, a member of the transforming growth factor (TGF)-β family. LEFTY2 is highly expressed in decidualizing human endometrial stromal cells (HESCs) during the late luteal phase of the menstrual cycle, coinciding with the closure of the window of implantation. Here, we show that flushing of the uterine lumen in mice with recombinant LEFTY2 inhibits the expression of key receptivity genes, including Cox2, Bmp2, and Wnt4, and blocks embryo implantation. In Ishikawa cells, a human endometrial epithelial cell line, LEFTY2 downregulated the expression of calcium release-activated calcium channel protein 1, encoded by ORAI1, and inhibited store-operated Ca 2+ entry (SOCE). Furthermore, LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, as well as YM-58483, inhibited, whereas the Ca 2+ ionophore, ionomycin, strongly upregulated COX2, BMP2 and WNT4 expression in decidualizing HESCs. These findings suggest that LEFTY2 closes the implantation window, at least in part, by downregulating Orai1, which in turn limits SOCE and antagonizes expression of Ca 2+-sensitive receptivity genes.

          Key messages

          •Endometrial receptivity is negatively regulated by LEFTY2.

          •LEFTY2 inhibits the expression of key murine receptivity genes, including Cox2, Bmp2 and Wnt4, and blocks embryo implantation.

          •LEFTY2 downregulates the expression of Orai1 and inhibits SOCE.

          •LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, and YM-58483 inhibit COX2, BMP2, and WNT4 expression in endometrial cells.

          •Targeting LEFTY2 and Orai1 may represent a novel approach for treating unexplained infertility.

          Electronic supplementary material

          The online version of this article (10.1007/s00109-017-1610-9) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references70

          • Record: found
          • Abstract: found
          • Article: not found

          Mitochondria and apoptosis.

          D Green, J Reed (1998)
          A variety of key events in apoptosis focus on mitochondria, including the release of caspase activators (such as cytochrome c), changes in electron transport, loss of mitochondrial transmembrane potential, altered cellular oxidation-reduction, and participation of pro- and antiapoptotic Bcl-2 family proteins. The different signals that converge on mitochondria to trigger or inhibit these events and their downstream effects delineate several major pathways in physiological cell death.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Store-Operated Calcium Channels.

            Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cyclic decidualization of the human endometrium in reproductive health and failure.

              Decidualization denotes the transformation of endometrial stromal fibroblasts into specialized secretory decidual cells that provide a nutritive and immunoprivileged matrix essential for embryo implantation and placental development. In contrast to most mammals, decidualization of the human endometrium does not require embryo implantation. Instead, this process is driven by the postovulatory rise in progesterone levels and increasing local cAMP production. In response to falling progesterone levels, spontaneous decidualization causes menstrual shedding and cyclic regeneration of the endometrium. A growing body of evidence indicates that the shift from embryonic to maternal control of the decidual process represents a pivotal evolutionary adaptation to the challenge posed by invasive and chromosomally diverse human embryos. This concept is predicated on the ability of decidualizing stromal cells to respond to individual embryos in a manner that either promotes implantation and further development or facilitates early rejection. Furthermore, menstruation and cyclic regeneration involves stem cell recruitment and renders the endometrium intrinsically capable of adapting its decidual response to maximize reproductive success. Here we review the endocrine, paracrine, and autocrine cues that tightly govern this differentiation process. In response to activation of various signaling pathways and genome-wide chromatin remodeling, evolutionarily conserved transcriptional factors gain access to the decidua-specific regulatory circuitry. Once initiated, the decidual process is poised to transit through distinct phenotypic phases that underpin endometrial receptivity, embryo selection, and, ultimately, resolution of pregnancy. We discuss how disorders that subvert the programming, initiation, or progression of decidualization compromise reproductive health and predispose for pregnancy failure.
                Bookmark

                Author and article information

                Contributors
                44 (0) 24 7696 8704 , J.J.Brosens@warwick.ac.uk
                +49 7071 29 72194 , florian.lang@uni-tuebingen.de
                Journal
                J Mol Med (Berl)
                J. Mol. Med
                Journal of Molecular Medicine (Berlin, Germany)
                Springer Berlin Heidelberg (Berlin/Heidelberg )
                0946-2716
                1432-1440
                11 December 2017
                11 December 2017
                2018
                : 96
                : 2
                : 173-182
                Affiliations
                [1 ]ISNI 0000 0001 2190 1447, GRID grid.10392.39, Department of Women’s Health, , Eberhard-Karls University Tuebingen, ; Calwerstr 7, D-72076 Tuebingen, Germany
                [2 ]ISNI 0000 0001 2190 1447, GRID grid.10392.39, Department of Medical Genetics and Applied Genomics, , Eberhard-Karls University Tuebingen, ; Calwerstr 7, D-72076 Tübingen, Germany
                [3 ]ISNI 0000 0000 8809 1613, GRID grid.7372.1, Division of Biomedical Sciences, , Warwick Medical School, ; Coventry, CV2 2DX UK
                [4 ]ISNI 0000 0001 2190 1447, GRID grid.10392.39, Department of Physiology I, , Eberhard-Karls University Tuebingen, ; Wilhelmstr 56, D-72074 Tuebingen, Germany
                [5 ]ISNI 0000 0001 2113 8111, GRID grid.7445.2, Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, , Imperial College London, ; London, W12 0NN UK
                [6 ]ISNI 0000 0001 2113 8111, GRID grid.7445.2, ES Cell and Transgenics Facility, Medical Research Council Clinical Sciences Centre, , Imperial College London, ; Du Cane Road, London, W12 0NN UK
                [7 ]ISNI 0000 0004 1936 8331, GRID grid.410356.5, Department of Biomedical and Molecular Sciences, , Queen’s University, ; Kingston, Ontario Canada
                [8 ]GRID grid.15628.38, Tommy’s National Centre for Miscarriage Research, , University Hospitals Coventry and Warwickshire NHS Trust, ; Clifford Bridge Rd, Coventry, CV2 2DX UK
                [9 ]ISNI 0000 0001 2176 9917, GRID grid.411327.2, Department of Molecular Medicine II, , Heinrich Heine University Düsseldorf, ; Universitätsstr 1, D-40225 Düsseldorf, Germany
                Article
                1610
                10.1007/s00109-017-1610-9
                5778154
                29230527
                9d8001c3-90ac-4e03-af39-bc1cad805fa5
                © The Author(s) 2017

                Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

                History
                : 3 May 2017
                : 16 October 2017
                : 2 November 2017
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100001659, Deutsche Forschungsgemeinschaft;
                Award ID: GRK 1302, SFB 773 B4/A1, La 315/13-3
                Award ID: zuk63
                Funded by: Warwick Medical School (GB)
                Funded by: FundRef http://dx.doi.org/10.13039/100004410, European Molecular Biology Organization;
                Award ID: ALTF 20-2013
                Funded by: FundRef http://dx.doi.org/10.13039/501100002345, Eberhard Karls Universität Tübingen;
                Award ID: Förtune 2426-0-0
                Award ID: open access fund
                Categories
                Original Article
                Custom metadata
                © Springer-Verlag GmbH Germany, part of Springer Nature 2018

                Molecular medicine
                implantation,lefty2,orai1,soce,calcium,pregnancy,miscarriage
                Molecular medicine
                implantation, lefty2, orai1, soce, calcium, pregnancy, miscarriage

                Comments

                Comment on this article