18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Follistatin‐like 1 promotes cardiac fibroblast activation and protects the heart from rupture

      research-article

      Read this article at

      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

          Follistatin‐like 1 (Fstl1) is a secreted protein that is acutely induced in heart following myocardial infarction ( MI). In this study, we investigated cell type‐specific regulation of Fstl1 and its function in a murine model of MI. Fstl1 was robustly expressed in fibroblasts and myofibroblasts in the infarcted area compared to cardiac myocytes. The conditional ablation of Fstl1 in S100a4‐expressing fibroblast lineage cells (Fstl1‐cf KO mice) led to a reduction in injury‐induced Fstl1 expression and increased mortality due to cardiac rupture during the acute phase. Cardiac rupture was associated with a diminished number of myofibroblasts and decreased expression of extracellular matrix proteins. The infarcts of Fstl1‐cf KO mice displayed weaker birefringence, indicative of thin and loosely packed collagen. Mechanistically, the migratory and proliferative capabilities of cardiac fibroblasts were attenuated by endogenous Fstl1 ablation. The activation of cardiac fibroblasts by Fstl1 was mediated by ERK1/2 but not Smad2/3 signaling. This study reveals that Fstl1 is essential for the acute repair of the infarcted myocardium and that stimulation of early fibroblast activation is a novel function of Fstl1.

          Related collections

          Most cited references44

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

          Identification and characterization of a fibroblast marker: FSP1

          We performed subtractive and differential hybridization for transcript comparison between murine fibroblasts and isogenic epithelium, and observed only a few novel intracellular genes which were relatively specific for fibroblasts. One such gene encodes a filament-associated, calcium-binding protein, fibroblast-specific protein 1 (FSP1). The promoter/enhancer region driving this gene is active in fibroblasts but not in epithelium, mesangial cells or embryonic endoderm. During development, FSP1 is first detected by in situ hybridization after day 8.5 as a postgastrulation event, and is associated with cells of mesenchymal origin or of fibroblastic phenotype. Polyclonal antiserum raised to recombinant FSP1 protein stained the cytoplasm of fibroblasts, but not epithelium. Only occasional cells stain with specific anti-FSP1 antibodies in normal parenchymal tissue. However, in kidneys fibrosing from persistent inflammation, many fibroblasts could be identified in interstitial sites of collagen deposition and also in tubular epithelium adjacent to the inflammatory process. This pattern of anti-FSP1 staining during tissue fibrosis suggests, as a hypothesis, that fibroblasts in some cases arise, as needed, from the local conversion of epithelium. Consistent with this notion that FSP1 may be involved in the transition from epithelium to fibroblasts are experiments in which the in vitro overexpression of FSP1 cDNA in tubular epithelium is accompanied by conversion to a mesenchymal phenotype, as characterized by a more stellate and elongated fibroblast- like appearance, a reduction in cytokeratin, and new expression of vimentin. Similarly, tubular epithelium submerged in type I collagen gels exhibited the conversion to a fibroblast phenotype which includes de novo expression of FSP1 and vimentin. Use of the FSP1 marker, therefore, should further facilitate both the in vivo studies of fibrogenesis and the mapping of cell fate among fibroblasts.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Epicardial FSTL1 reconstitution regenerates the adult mammalian heart.

            The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              GDF-15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice.

              Inflammatory cell recruitment after myocardial infarction needs to be tightly controlled to permit infarct healing while avoiding fatal complications such as cardiac rupture. Growth differentiation factor-15 (GDF-15), a transforming growth factor-β (TGF-β)-related cytokine, is induced in the infarcted heart of mice and humans. We show that coronary artery ligation in Gdf15-deficient mice led to enhanced recruitment of polymorphonuclear leukocytes (PMNs) into the infarcted myocardium and an increased incidence of cardiac rupture. Conversely, infusion of recombinant GDF-15 repressed PMN recruitment after myocardial infarction. In vitro, GDF-15 inhibited PMN adhesion, arrest under flow and transendothelial migration. Mechanistically, GDF-15 counteracted chemokine-triggered conformational activation and clustering of β(2) integrins on PMNs by activating the small GTPase Cdc42 and inhibiting activation of the small GTPase Rap1. Intravital microscopy in vivo in Gdf15-deficient mice showed that Gdf-15 is required to prevent excessive chemokine-activated leukocyte arrest on the endothelium. Genetic ablation of β(2) integrins in myeloid cells rescued the mortality of Gdf15-deficient mice after myocardial infarction. To our knowledge, GDF-15 is the first cytokine identified as an inhibitor of PMN recruitment by direct interference with chemokine signaling and integrin activation. Loss of this anti-inflammatory mechanism leads to fatal cardiac rupture after myocardial infarction.
                Bookmark

                Author and article information

                Journal
                EMBO Mol Med
                EMBO Mol Med
                10.1002/(ISSN)1757-4684
                EMMM
                embomm
                EMBO Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1757-4676
                1757-4684
                27 May 2016
                August 2016
                : 8
                : 8 ( doiID: 10.1002/emmm.v8.8 )
                : 949-966
                Affiliations
                [ 1 ] Department of Molecular Cardiology Whitaker Cardiovascular InstituteBoston University School of Medicine Boston MAUSA
                [ 2 ] Department of Anatomy, Embryology and PhysiologyAcademic Medical Center AmsterdamThe Netherlands
                [ 3 ] Molecular Cardiovascular MedicineNagoya University Graduate School of Medicine Showa‐ku, NagoyaJapan
                [ 4 ] Cardiovascular Research CenterLewis Katz School of Medicine at Temple University Philadelphia PAUSA
                [ 5 ] Institute of Life SciencesScuola Superiore Sant'Anna PisaItaly
                Author notes
                [*] [* ]Corresponding author. Tel: +1 617 414 2390; Fax: +1 617 414 2391; E‐mail: kxwalsh@ 123456bu.edu
                Author information
                http://orcid.org/0000-0001-7580-2276
                Article
                EMMM201506151
                10.15252/emmm.201506151
                4967946
                27234440
                27e26114-5c6a-4af8-97fb-4e1760c63623
                © 2016 The Authors. Published under the terms of the CC BY 4.0 license

                This is an open access article under the terms of the Creative Commons Attribution 4.0 License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 15 December 2015
                : 22 April 2016
                : 27 April 2016
                Page count
                Pages: 18
                Funding
                Funded by: NIH
                Award ID: HL081587
                Award ID: HL116591
                Award ID: HL120160
                Award ID: HL126141
                Award ID: HL74237
                Award ID: HL108213
                Funded by: Takeda Pharmaceuticals
                Funded by: Postdoctoral Research Fellowship in Cardiology by Japan Heart Foundation and Bayer (Japan)
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                emmm201506151
                August 2016
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.9.4 mode:remove_FC converted:01.08.2016

                Molecular medicine
                cardiokine,fibrosis,infarct healing,myocardial infarction,cardiovascular system
                Molecular medicine
                cardiokine, fibrosis, infarct healing, myocardial infarction, cardiovascular system

                Comments

                Comment on this article