21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Transcription factor KLF6 upregulates expression of metalloprotease MMP14 and subsequent release of soluble endoglin during vascular injury

      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

          After endothelial injury, the transcription factor Krüppel-like factor 6 (KLF6) translocates into the cell nucleus to regulate a variety of target genes involved in angiogenesis, vascular repair and remodeling, including components of the membrane transforming growth factor beta (TGF-β) receptor complex such as endoglin and activin receptor-like kinase 1. The membrane metalloproteinase 14 (MMP14 or MT1-MMP) targets endoglin to release soluble endoglin and is involved in vascular inflammation and endothelial tubulogenesis. However, little is known about the regulation of MMP14 expression during vascular wounding. In vitro denudation of monolayers of human endothelial cell monolayers leads to an increase in the KLF6 gene transcriptional rate, followed by an upregulation of MMP14 and release of soluble endoglin. Concomitant with this process, MMP14 co-localizes with endoglin in the sprouting endothelial cells surrounding the wound border. MMP14 expression at mRNA and protein levels is increased by ectopic KLF6 and downregulated by KLF6 suppression in cultured endothelial cells. Moreover, after wire-induced endothelial denudation, Klf6 +/− mice show lower levels of MMP14 in their vasculature compared with their wild-type siblings. Ectopic cellular expression of KLF6 results in an increased transcription rate of MMP14, and chromatin immunoprecipitation assays show that KLF6 interacts with MMP14 promoter in ECs, this interaction being enhanced during wound healing. Furthermore, KLF6 markedly increases the transcriptional activity of different reporter constructs of MMP14 gene promoter. These results suggest that KLF6 regulates MMP14 transcription and is a critical player of the gene expression network triggered during endothelial repair.

          Electronic supplementary material

          The online version of this article (doi:10.1007/s10456-016-9495-8) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references42

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

          Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization.

          The extracellular matrix (ECM) is critical for all aspects of vascular biology. In concert with supporting cells, endothelial cells (ECs) assemble a laminin-rich basement membrane matrix that provides structural and organizational stability. During the onset of angiogenesis, this basement membrane matrix is degraded by proteinases, among which membrane-type matrix metalloproteinases (MT-MMPs) are particularly significant. As angiogenesis proceeds, ECM serves essential functions in supporting key signaling events involved in regulating EC migration, invasion, proliferation, and survival. Moreover, the provisional ECM serves as a pliable scaffold wherein mechanical guidance forces are established among distal ECs, thereby providing organizational cues in the absence of cell-cell contact. Finally, through specific integrin-dependent signal transduction pathways, ECM controls the EC cytoskeleton to orchestrate the complex process of vascular morphogenesis by which proliferating ECs organize into multicellular tubes with functional lumens. Thus, the composition of ECM and therefore the regulation of ECM degradation and remodeling serves pivotally in the control of lumen and tube formation and, finally, neovessel stability and maturation.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Extracellular control of TGFbeta signalling in vascular development and disease.

            The intracellular mechanism of transforming growth factor-beta (TGFbeta) signalling via kinase receptors and SMAD effectors is firmly established, but recent studies of human cardiovascular syndromes such as Marfan syndrome and pre-eclampsia have refocused attention on the importance of regulating the availability of active extracellular TGFbeta. It seems that elastic extracellular matrix (ECM) components have a crucial role in controlling TGFbeta signalling, while soluble and membrane bound forms of TGFbeta co-receptors add further layers of regulation. Together, these extracellular interactions determine the final bioavailability of TGFbeta to vascular cells, and dysregulation is associated with an increasing number of vascular pathologies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Matrix metalloproteinases and angiogenesis.

              Matrix metalloproteinases (MMPs) are a family of enzymes that proteolytically degrade various components of the extracellular matrix (ECM). Angiogenesis is the process of forming new blood vessels from existing ones and requires degradation of the vascular basement membrane and remodeling of the ECM in order to allow endothelial cells to migrate and invade into the surrounding tissue. MMPs participate in this remodeling of basement membranes and ECM. However, it has become clear that MMPs contribute more to angiogenesis than just degrading ECM components. Specific MMPs have been shown to enhance angiogenesis by helping to detach pericytes from vessels undergoing angiogenesis, by releasing ECM-bound angiogenic growth factors, by exposing cryptic proangiogenic integrin binding sites in the ECM, by generating promigratory ECM component fragments, and by cleaving endothelial cell-cell adhesions. MMPs can also contribute negatively to angiogenesis through the generation of endogenous angiogenesis inhibitors by proteolytic cleavage of certain collagen chains and plasminogen and by modulating cell receptor signaling by cleaving off their ligand-binding domains. A number of inhibitors of MMPs that show antiangiogenic activity are already in early stages of clinical trials, primarily to treat cancer and cancer-associated angiogenesis. However, because of the multiple effects of MMPs on angiogenesis, careful testing of these MMP inhibitors is necessary to show that these compounds do not actually enhance angiogenesis.
                Bookmark

                Author and article information

                Contributors
                34-918373112 , bernabeu.c@cib.csic.es
                Journal
                Angiogenesis
                Angiogenesis
                Angiogenesis
                Springer Netherlands (Dordrecht )
                0969-6970
                1573-7209
                5 February 2016
                5 February 2016
                2016
                : 19
                : 155-171
                Affiliations
                [ ]Centro de Investigaciones Biológicas, CSIC, c/Ramiro de Maeztu 9, Madrid, 28040 Spain
                [ ]Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, 28040 Spain
                [ ]Servei de Cardiologia, Hospital Clínic i Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
                [ ]Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY USA
                [ ]Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
                [ ]National Institute for Health Research, Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
                Article
                9495
                10.1007/s10456-016-9495-8
                4819519
                26850053
                8354b605-d742-4360-a2ae-8ae6c9085dcb
                © The Author(s) 2016

                Open AccessThis 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
                : 30 August 2015
                : 23 January 2016
                Funding
                Funded by: Ministerio de Economia y Competitividad of Spain
                Award ID: SAF2013-43421-R
                Award ID: SAF2011-23475
                Award Recipient :
                Funded by: Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)
                Award ID: ISCIII-CB06/07/0038
                Award ID: ER16PIAC707
                Award Recipient :
                Funded by: Red de Investigación Cooperativa en Salud, Spain
                Award ID: RD12/0042/0006
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health (US);
                Award ID: DK37340 and DK56621
                Award Recipient :
                Categories
                Original Paper
                Custom metadata
                © Springer Science+Business Media Dordrecht 2016

                Human biology
                endothelial cells,vascular injury,transcription,klf6,mmp14,soluble endoglin
                Human biology
                endothelial cells, vascular injury, transcription, klf6, mmp14, soluble endoglin

                Comments

                Comment on this article