15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      A Mosaic Mouse Model of Astrocytoma Identifies αvβ8 Integrin as an Essential Regulator of Tumor-Induced Angiogenesis

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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

          The process of angiogenesis involves a complex set of cell-cell and cell-extracellular matrix (ECM) interactions that coordinately regulate new blood vessel growth and maturation. Although many factors that promote angiogenesis have been characterized, the identities and mechanisms of action of many endogenous inhibitors of angiogenesis remain unclear. Furthermore, little is known about how tumor cells selectively circumvent the actions of these inhibitors to drive pathological angiogenesis, a requisite event for tumor progression. Using mosaic mouse models of the malignant brain cancer, astrocytoma, we report that tumor cells induce pathological angiogenesis by suppressing expression of the ECM protein receptor αvβ8 integrin. Diminished integrin expression in astrocytomas cells leads to reduced activation of latent TGFβs, resulting in impaired TGFβ receptor signaling events in tumor-associated endothelial cells. These data reveal that astrocytoma cells manipulate their angiogenic balance by selectively suppressing αvβ8 integrin expression/function, and also demonstrate that an adhesion and signaling axis normally involved in developmental brain angiogenesis is pathologically exploited in adult brain tumors.

          Related collections

          Most cited references34

          • 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

            P53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation

            Glioblastoma (GBM) is a highly lethal brain tumor presenting as one of two subtypes with distinct clinical histories and molecular profiles. The primary GBM subtype presents acutely as high-grade disease that typically harbors EGFR, Pten and Ink4a/Arf mutations, and the secondary GBM subtype evolves from the slow progression of low-grade disease that classically possesses PDGF and p53 events 1–3 . Here, we show that concomitant CNS-specific deletion of p53 and Pten in the mouse CNS generates a penetrant acute-onset high-grade malignant glioma phenotype with striking clinical, pathological and molecular resemblance to primary GBM in humans. This genetic observation prompted p53 and Pten mutational analysis in human primary GBM, demonstrating unexpectedly frequent inactivating mutations of p53 as well the expected Pten mutations. Integrated transcriptomic profiling, in silico promoter analysis and functional studies of murine neural stem cells (NSCs) established that dual, but not singular, inactivation of p53 and Pten promotes an undifferentiated state with high renewal potential and drives elevated c-Myc levels and its associated signature. Functional studies validated increased c-Myc activity as a potent contributor to the impaired differentiation and enhanced renewal of p53-Pten null NSCs as well as tumor neurospheres (TNSs) derived from this model. c-Myc also serves to maintain robust tumorigenic potential of p53-Pten null TNSs. These murine modeling studies, together with confirmatory transcriptomic/promoter studies in human primary GBM, validate a pathogenetic role of a common tumor suppressor mutation profile in human primary GBM and establish c-Myc as a key target for cooperative actions of p53 and Pten in the regulation of normal and malignant stem/progenitor cell differentiation, self-renewal and tumorigenic potential.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model.

              Malignant astrocytomas are infiltrative and incurable brain tumors. Despite profound therapeutic implications, the identity of the cell (or cells) of origin has not been rigorously determined. We previously reported mouse models based on conditional inactivation of the human astrocytoma-relevant tumor suppressors p53, Nf1, and Pten, wherein through somatic loss of heterozygosity, mutant mice develop tumors with 100% penetrance. In the present study, we show that tumor suppressor inactivation in neural stem/progenitor cells is both necessary and sufficient to induce astrocytoma formation. We demonstrate in vivo that transformed cells and their progeny undergo infiltration and multilineage differentiation during tumorigenesis. Tumor suppressor heterozygous neural stem/progenitor cultures from presymptomatic mice show aberrant growth advantage and altered differentiation, thus identifying a pretumorigenic cell population.
                Bookmark

                Author and article information

                Journal
                8711562
                6325
                Oncogene
                Oncogene
                Oncogene
                0950-9232
                1476-5594
                27 May 2010
                07 June 2010
                5 August 2010
                11 February 2011
                : 29
                : 31
                : 4460-4472
                Affiliations
                []Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, USA, 77030
                [¥ ]Department of Pathology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, USA, 77030
                Author notes
                [* ]Corresponding author: Department of Cancer Biology, Unit 173 University of Texas, M.D. Anderson Cancer Center 1515 Holcombe Boulevard Houston, TX 77030 jhmccarty@ 123456mdanderson.org
                Article
                NIHMS198457
                10.1038/onc.2010.199
                3037767
                20531304
                d393b171-1ed2-41a3-9a6c-594b9bb52097

                Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Funding
                Funded by: National Institute of Neurological Disorders and Stroke : NINDS
                Award ID: R01 NS059876-03 || NS
                Funded by: National Institute of Neurological Disorders and Stroke : NINDS
                Award ID: R01 NS059876-02S2 || NS
                Categories
                Article

                Oncology & Radiotherapy
                tgfβ,glioblastoma multiforme,glioma,extracellular matrix,astrocyte,itgb8
                Oncology & Radiotherapy
                tgfβ, glioblastoma multiforme, glioma, extracellular matrix, astrocyte, itgb8

                Comments

                Comment on this article