29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extracellular vesicles

      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 multiforme (GBM) are mortifying brain tumours that contain a subpopulation of tumour cells with stem-like properties, termed glioblastoma stem-like cells (GSCs). GSCs largely contribute to tumour initiation, propagation and resistance to current anti-cancer therapies. GSCs are situated in perivascular niches, closely associated with brain microvascular endothelial cells, thereby involved in bidirectional molecular and cellular interactions. Moreover, extracellular vesicles are suspected to carry essential information that can adapt the microenvironment to the tumour’s needs, including tumour-induced angiogenesis. In GBM, extracellular vesicles produced by differentiated tumour cells and GSCs were demonstrated to disseminate locally and at distance. Here, we report that the pro-angiogenic pro-permeability factor VEGF-A is carried in extracellular vesicles secreted from ex vivo cultured patient-derived GSCs. Of note, extracellular vesicle-derived VEGF-A contributes to the in vitro elevation of permeability and angiogenic potential in human brain endothelial cells. Indeed, VEGF-A silencing in GSCs compromised in vitro extracellular vesicle-mediated increase in permeability and angiogenesis. From a clinical standpoint, extracellular vesicles isolated from circulating blood of GBM patients present higher levels of VEGF-A, as compared to healthy donors. Overall, our results suggest that extracellular vesicle-harboured VEGF-A targets brain endothelial cells and might impact their ability to form new vessels. Thus, tumour-released EV cargo might emerge as an instrumental part of the tumour-induced angiogenesis and vascular permeability modus operandi in GBM.

          Related collections

          Most cited references45

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

          Evolution of the cancer stem cell model.

          Genetic analyses have shaped much of our understanding of cancer. However, it is becoming increasingly clear that cancer cells display features of normal tissue organization, where cancer stem cells (CSCs) can drive tumor growth. Although often considered as mutually exclusive models to describe tumor heterogeneity, we propose that the genetic and CSC models of cancer can be harmonized by considering the role of genetic diversity and nongenetic influences in contributing to tumor heterogeneity. We offer an approach to integrating CSCs and cancer genetic data that will guide the field in interpreting past observations and designing future studies. Copyright © 2014 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Protein typing of circulating microvesicles allows real-time monitoring of glioblastoma therapy.

            Glioblastomas shed large quantities of small, membrane-bound microvesicles into the circulation. Although these hold promise as potential biomarkers of therapeutic response, their identification and quantification remain challenging. Here, we describe a highly sensitive and rapid analytical technique for profiling circulating microvesicles directly from blood samples of patients with glioblastoma. Microvesicles, introduced onto a dedicated microfluidic chip, are labeled with target-specific magnetic nanoparticles and detected by a miniaturized nuclear magnetic resonance system. Compared with current methods, this integrated system has a much higher detection sensitivity and can differentiate glioblastoma multiforme (GBM) microvesicles from nontumor host cell-derived microvesicles. We also show that circulating GBM microvesicles can be used to analyze primary tumor mutations and as a predictive metric of treatment-induced changes. This platform could provide both an early indicator of drug efficacy and a potential molecular stratifier for human clinical trials.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Extracellular vesicles from neural stem cells transfer IFN-γ via Ifngr1 to activate Stat1 signaling in target cells.

              The idea that stem cell therapies work only via cell replacement is challenged by the observation of consistent intercellular molecule exchange between the graft and the host. Here we defined a mechanism of cellular signaling by which neural stem/precursor cells (NPCs) communicate with the microenvironment via extracellular vesicles (EVs), and we elucidated its molecular signature and function. We observed cytokine-regulated pathways that sort proteins and mRNAs into EVs. We described induction of interferon gamma (IFN-γ) pathway in NPCs exposed to proinflammatory cytokines that is mirrored in EVs. We showed that IFN-γ bound to EVs through Ifngr1 activates Stat1 in target cells. Finally, we demonstrated that endogenous Stat1 and Ifngr1 in target cells are indispensable to sustain the activation of Stat1 signaling by EV-associated IFN-γ/Ifngr1 complexes. Our study identifies a mechanism of cellular signaling regulated by EV-associated IFN-γ/Ifngr1 complexes, which grafted stem cells may use to communicate with the host immune system.
                Bookmark

                Author and article information

                Journal
                J Extracell Vesicles
                J Extracell Vesicles
                ZJEV
                zjev20
                Journal of Extracellular Vesicles
                Taylor & Francis
                2001-3078
                2017
                8 August 2017
                : 6
                : 1
                : 1359479
                Affiliations
                [ a ] CNRS, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Institut Cochin , Paris, France
                [ b ] INSERM, CNRS, CRCINA, Team SOAP, Université de Nantes , Nantes, France
                [ c ] Hôpital d’Instruction des Armées Percy, Service de Santé des Armées , Clamart, France
                [ d ] Ecole du Val-de-Grâce, Service de Santé des Armées , Paris, France
                [ e ] CNRS, UMR 8257 , Paris, France
                Author notes
                CONTACT Julie Gavard julie.gavard@ 123456inserm.fr Team SOAP, Signaling in Oncogenesis, Angiogenesis and Permeability, Cancer Research Center Nantes-Angers , IRS-UN blg, Room 416, 8 quai Moncousu, Nantes 44000, France
                Author information
                http://orcid.org/0000-0003-0735-9000
                http://orcid.org/0000-0002-7985-9007
                Article
                1359479
                10.1080/20013078.2017.1359479
                5549846
                28815003
                fb9bf902-8429-4a97-9aa2-e1276fc9f4d1
                © 2017 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 29 December 2016
                : 20 July 2017
                Page count
                Figures: 5, References: 74, Pages: 13
                Funding
                Funded by: Fondation ARC pour la Recherche sur le Cancer 10.13039/501100004097
                Award ID: PJA 20151203221
                Funded by: Fondation ARC pour la Recherche sur le Cancer 10.13039/501100004097
                Award ID: M2R20160604052
                Funded by: Ligue Contre le Cancer 10.13039/501100004099
                Award ID: Comite Loire Atlantique
                Award ID: Vendée
                Award ID: Morbihan
                Funded by: Nantes Metropole (FR)
                Award ID: Connect Talent
                Funded by: Region Pays de La Loire (FR)
                Award ID: Connect Talent
                This work was supported by the Fondation ARC pour la Recherche sur le Cancer [PJA 20151203221]; Fondation ARC pour la Recherche sur le Cancer [M2R20160604052]; Fondation Bettencourt Schueller; Fondation pour la Recherche Médicale; Institut National Du Cancer; Ligue Contre le Cancer [Comite Loire Atlantique, Vendée, Morbihan]; Nantes Metropole (FR) [Connect Talent]; Region Pays de La Loire (FR) [Connect Talent]; Université Paris Descartes;
                Categories
                Research Article
                Research Article

                angiogenesis,glioma,gsc,cancer stem-like cells,vegf,exosomes,microvesicles,microparticles,circulating vesicles

                Comments

                Comment on this article