11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Interleukin-8 Secreted by Glioblastoma Cells Induces Microvascular Hyperpermeability Through NO Signaling Involving S-Nitrosylation of VE-Cadherin and p120 in Endothelial Cells

      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

          Glioblastoma is a highly aggressive brain tumor, characterized by the formation of dysfunctional blood vessels and a permeable endothelial barrier. S-nitrosylation, a post-translational modification, has been identified as a regulator of endothelial function. In this work we explored whether S-nitrosylation induced by glioblastoma tumors regulates the endothelial function. As proof of concept, we observed that S-nitrosylation is present in the tumoral microenvironment of glioblastoma in two different animal models. Subsequently, we measured S nitrosylation and microvascular permeability in EAhy296 endothelial cells and in cremaster muscle. In vitro, conditioned medium from the human glioblastoma cell line U87 activates endothelial nitric oxide synthase, causes VE-cadherin- S-nitrosylation and induces hyperpermeability. Blocking Interleukin-8 (IL-8) in the conditioned medium inhibited S-nitrosylation of VE-cadherin and hyperpermeability. Recombinant IL-8 increased endothelial permeability by activating eNOS, S-nitrosylation of VE-cadherin and p120, internalization of VE-cadherin and disassembly of adherens junctions. In vivo, IL-8 induced S-nitrosylation of VE-cadherin and p120 and conditioned medium from U87 cells caused hyperpermeability in the mouse cremaster muscle. We conclude that eNOS signaling induced by glioma cells-secreted IL-8 regulates endothelial barrier function in the context of glioblastoma involving S-nitrosylation of VE-cadherin and p120. Our results suggest that inhibiting S-nitrosylation may be an effective way to control and/or block damage to the endothelial barrier and prevent cancer progression.

          Related collections

          Most cited references51

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

          PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.

          Mapping of homozygous deletions on human chromosome 10q23 has led to the isolation of a candidate tumor suppressor gene, PTEN, that appears to be mutated at considerable frequency in human cancers. In preliminary screens, mutations of PTEN were detected in 31% (13/42) of glioblastoma cell lines and xenografts, 100% (4/4) of prostate cancer cell lines, 6% (4/65) of breast cancer cell lines and xenografts, and 17% (3/18) of primary glioblastomas. The predicted PTEN product has a protein tyrosine phosphatase domain and extensive homology to tensin, a protein that interacts with actin filaments at focal adhesions. These homologies suggest that PTEN may suppress tumor cell growth by antagonizing protein tyrosine kinases and may regulate tumor cell invasion and metastasis through interactions at focal adhesions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Angiogenesis in brain tumours.

            Despite aggressive surgery, radiotherapy and chemotherapy, malignant gliomas remain uniformly fatal. To progress, these tumours stimulate the formation of new blood vessels through processes driven primarily by vascular endothelial growth factor (VEGF). However, the resulting vessels are structurally and functionally abnormal, and contribute to a hostile microenvironment (low oxygen tension and high interstitial fluid pressure) that selects for a more malignant phenotype with increased morbidity and mortality. Emerging preclinical and clinical data indicate that anti-VEGF therapies are potentially effective in glioblastoma--the most frequent primary brain tumour--and can transiently normalize tumour vessels. This creates a window of opportunity for optimally combining chemotherapeutics and radiation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Permanent cell line expressing human factor VIII-related antigen established by hybridization.

              A permanent human cell line, EA . hy 926, has been established that expresses at least one highly differentiated function of vascular endothelium, factor VIII-related antigen. This line was derived by fusing human umbilical vein endothelial cells with the permanent human cell line A549. Hybrid cells that survived in selective medium had more chromosomes than either progenitor cell type and included a marker chromosome from the A549 line. Factor VIII-related antigen can be identified intracellularly in the hybrids by immunofluorescence and accumulates in the culture fluid. Expression of factor VIII-related antigen by these hybrid cells has been maintained for more than 100 cumulative population doublings, including more than 50 passages and three cloning steps. This is evidence that EA . hy 926 represents a permanent line.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Physiol
                Front Physiol
                Front. Physiol.
                Frontiers in Physiology
                Frontiers Media S.A.
                1664-042X
                08 August 2019
                2019
                : 10
                : 988
                Affiliations
                [1] 1Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile , Valdivia, Chile
                [2] 2Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile , Valdivia, Chile
                [3] 3Instituto de Histología, Anatomía y Patología, Facultad de Medicina, Universidad Austral de Chile , Valdivia, Chile
                [4] 4Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile , Valdivia, Chile
                [5] 5Departamento de Fisiología, Pontificia Universidad Católica de Chile , Santiago, Chile
                [6] 6Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile , Santiago, Chile
                [7] 7Team SOAP, Signaling in Oncogenesis, Angiogenesis and Permeability, INSERM, CNRS, Institut de Cancérologie de l’Ouest, Université de Nantes , Nantes, France
                [8] 8Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, The State University of New Jersey , Newark, NJ, United States
                [9] 9Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile , Valdivia, Chile
                Author notes

                Edited by: Claudia Penna, University of Turin, Italy

                Reviewed by: Muhammad Aslam, University of Giessen, Germany; Bingmei Fu, The City College of New York (CUNY), United States

                *Correspondence: José Sarmiento, jsarmien@ 123456uach.cl
                Fabiola A. Sánchez, fabiolasanchez@ 123456uach.cl

                This article was submitted to Vascular Physiology, a section of the journal Frontiers in Physiology

                Article
                10.3389/fphys.2019.00988
                6694439
                31501653
                2f389ead-5ab5-45c6-8b23-3187a1069cea
                Copyright © 2019 Guequén, Zamorano, Córdova, Koning, Torres, Ehrenfeld, Boric, Salazar-Onfray, Gavard, Durán, Quezada, Sarmiento and Sánchez.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 05 March 2019
                : 18 July 2019
                Page count
                Figures: 6, Tables: 0, Equations: 0, References: 55, Pages: 12, Words: 0
                Funding
                Funded by: Fondo Nacional de Desarrollo Científico y Tecnológico 10.13039/501100002850
                Funded by: Dirección de Dirección, Universidad Austral de Chile 10.13039/501100007159
                Funded by: Foundation for the National Institutes of Health 10.13039/100000009
                Categories
                Physiology
                Original Research

                Anatomy & Physiology
                adherens junction,glioblastoma,endothelial permeability,s-nitrosylation,ve-cadherin

                Comments

                Comment on this article