24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      IFN- γ drives inflammatory bowel disease pathogenesis through VE-cadherin–directed vascular barrier disruption

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          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

          Inflammatory bowel disease (IBD) is a chronic inflammatory disorder with rising incidence. Diseased tissues are heavily vascularized. Surprisingly, the pathogenic impact of the vasculature in IBD and the underlying regulatory mechanisms remain largely unknown. IFN-γ is a major cytokine in IBD pathogenesis, but in the context of the disease, it is almost exclusively its immune-modulatory and epithelial cell–directed functions that have been considered. Recent studies by our group demonstrated that IFN-γ also exerts potent effects on blood vessels. Based on these considerations, we analyzed the vessel-directed pathogenic functions of IFN-γ and found that it drives IBD pathogenesis through vascular barrier disruption. Specifically, we show that inhibition of the IFN-γ response in vessels by endothelial-specific knockout of IFN-γ receptor 2 ameliorates experimentally induced colitis in mice. IFN-γ acts pathogenic by causing a breakdown of the vascular barrier through disruption of the adherens junction protein VE-cadherin. Notably, intestinal vascular barrier dysfunction was also confirmed in human IBD patients, supporting the clinical relevance of our findings. Treatment with imatinib restored VE-cadherin/adherens junctions, inhibited vascular permeability, and significantly reduced colonic inflammation in experimental colitis. Our findings inaugurate the pathogenic impact of IFN-γ–mediated intestinal vessel activation in IBD and open new avenues for vascular-directed treatment of this disease.

          Abstract

          Related collections

          Most cited references57

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

          Molecular regulation of vessel maturation.

          The maturation of nascent vasculature, formed by vasculogenesis or angiogenesis, requires recruitment of mural cells, generation of an extracellular matrix and specialization of the vessel wall for structural support and regulation of vessel function. In addition, the vascular network must be organized so that all the parenchymal cells receive adequate nutrients. All of these processes are orchestrated by physical forces as well as by a constellation of ligands and receptors whose spatio-temporal patterns of expression and concentration are tightly regulated. Inappropriate levels of these physical forces or molecules produce an abnormal vasculature--a hallmark of various pathologies. Normalization of the abnormal vasculature can facilitate drug delivery to tumors and formation of a mature vasculature can help realize the promise of therapeutic angiogenesis and tissue engineering.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The role of adherens junctions and VE-cadherin in the control of vascular permeability.

            Endothelial cells control the passage of plasma constituents and circulating cells from blood to the underlying tissues. This specialized function is lost or impaired in several pathological conditions - including inflammation, sepsis, ischemia and diabetes - which leads to severe, and sometimes fatal, organ dysfunction. Endothelial permeability is regulated in part by the dynamic opening and closure of cell-cell adherens junctions (AJs). In endothelial cells, AJs are largely composed of vascular endothelial cadherin (VE-cadherin), an endothelium-specific member of the cadherin family of adhesion proteins that binds, via its cytoplasmic domain, to several protein partners, including p120, beta-catenin and plakoglobin. Endogenous pathways that increase vascular permeability affect the function and organization of VE-cadherin and other proteins at AJs in diverse ways. For instance, several factors, including vascular endothelial growth factor (VEGF), induce the tyrosine phosphorylation of VE-cadherin, which accompanies an increase in vascular permeability and leukocyte diapedesis; in addition, the internalization and cleavage of VE-cadherin can cause AJs to be dismantled. From the knowledge of how AJ organization can be modulated, it is possible to formulate several pharmacological strategies to control the barrier function of the endothelium. We discuss the possible use of inhibitors of SRC and other kinases, of agents that increase cAMP levels, and of inhibitors of lytic enzymes as pharmacological tools for decreasing endothelial permeability.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              A review of activity indices and efficacy end points for clinical trials of medical therapy in adults with ulcerative colitis.

                Bookmark

                Author and article information

                Contributors
                Journal
                J Clin Invest
                J. Clin. Invest
                J Clin Invest
                The Journal of Clinical Investigation
                American Society for Clinical Investigation
                0021-9738
                1558-8238
                30 September 2019
                30 September 2019
                1 November 2019
                1 February 2020
                : 129
                : 11
                : 4691-4707
                Affiliations
                [1 ]Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Medical Center Erlangen,
                [2 ]Division of Genetics, Nikolaus-Fiebiger-Center of Molecular Medicine,
                [3 ]Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, University Medical Center Erlangen, and
                [4 ]Optical Imaging Centre, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
                [5 ]Program for Bioengineering, School of Engineering, Seoul National University, Seoul, Republic of Korea.
                [6 ]Department of Internal Medicine 3, Rheumatology and Immunology, University Medical Center Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
                [7 ]Medical Clinic I, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin, Germany.
                [8 ]Department of Surgery and
                [9 ]Institute of Pathology, University Medical Center Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
                [10 ]Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany.
                [11 ]Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
                Author notes
                Address correspondence to: Michael Stürzl, Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Medical Center Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 12, 91054 Erlangen, Germany. Phone: 49.9131.85.39522; Email: michael.stuerzl@ 123456uk-erlangen.de .
                Author information
                http://orcid.org/0000-0002-9327-2296
                http://orcid.org/0000-0001-9189-0384
                http://orcid.org/0000-0002-2124-3103
                http://orcid.org/0000-0002-0945-0660
                http://orcid.org/0000-0003-3031-7677
                http://orcid.org/0000-0003-1291-622X
                http://orcid.org/0000-0002-0494-7117
                http://orcid.org/0000-0002-9276-2824
                Article
                PMC6819119 PMC6819119 6819119 124884
                10.1172/JCI124884
                6819119
                31566580
                79f50095-bf90-492e-9480-d1d5a8548f59
                © 2019 American Society for Clinical Investigation
                History
                : 13 September 2018
                : 1 August 2019
                Funding
                Funded by: German Research Foundation (DFG)
                Award ID: FOR 2438 (subproject 2)
                Funded by: German Research Foundation (DFG)
                Award ID: KFO 257 (subproject 4)
                Funded by: German Research Foundation (DFG)
                Award ID: SFB/TRR241 (subproject A06)
                Funded by: German Research Foundation (DFG)
                Award ID: BR 5196/2-1
                Funded by: Interdisciplinary Center for Clinical Research (IZKF) of the Clinical Center Erlangen
                Award ID: D28
                Funded by: W. Lutz Stiftung
                Award ID: to MS
                Funded by: Forschungsstiftung Medizin am Universitätsklinikum Erlangen
                Award ID: to MS
                no number available
                no number available
                Categories
                Research Article

                Vascular Biology,endothelial cells,Inflammatory bowel disease,Cytokines,Gastroenterology

                Comments

                Comment on this article