2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PELP1 promotes glioblastoma progression by enhancing Wnt/β-catenin signaling

      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

          Background

          Glioblastoma (GBM) is a deadly neoplasm of the central nervous system. The molecular mechanisms and players that contribute to GBM development is incompletely understood.

          Methods

          The expression of PELP1 in different grades of glioma and normal brain tissues was analyzed using immunohistochemistry on a tumor tissue array. PELP1 expression in established and primary GBM cell lines was analyzed by Western blotting. The effect of PELP1 knockdown was studied using cell proliferation, colony formation, migration, and invasion assays. Mechanistic studies were conducted using RNA-seq, RT-qPCR, immunoprecipitation, reporter gene assays, and signaling analysis. Mouse orthotopic models were used for preclinical evaluation of PELP1 knock down.

          Results

          Nuclear receptor coregulator PELP1 is highly expressed in gliomas compared to normal brain tissues, with the highest expression in GBM. PELP1 expression was elevated in established and patient-derived GBM cell lines compared to normal astrocytes. Knockdown of PELP1 resulted in a significant decrease in cell viability, survival, migration, and invasion. Global RNA-sequencing studies demonstrated that PELP1 knockdown significantly reduced the expression of genes involved in the Wnt/β-catenin pathway. Mechanistic studies demonstrated that PELP1 interacts with and functions as a coactivator of β-catenin. Knockdown of PELP1 resulted in a significant increase in survival of mice implanted with U87 and GBM PDX models.

          Conclusions

          PELP1 expression is upregulated in GBM and PELP1 signaling via β-catenin axis contributes to GBM progression. Thus, PELP1 could be a potential target for the development of therapeutic intervention in GBM.

          Related collections

          Most cited references65

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

          Brain tumour cells interconnect to a functional and resistant network.

          Astrocytic brain tumours, including glioblastomas, are incurable neoplasms characterized by diffusely infiltrative growth. Here we show that many tumour cells in astrocytomas extend ultra-long membrane protrusions, and use these distinct tumour microtubes as routes for brain invasion, proliferation, and to interconnect over long distances. The resulting network allows multicellular communication through microtube-associated gap junctions. When damage to the network occurred, tumour microtubes were used for repair. Moreover, the microtube-connected astrocytoma cells, but not those remaining unconnected throughout tumour progression, were protected from cell death inflicted by radiotherapy. The neuronal growth-associated protein 43 was important for microtube formation and function, and drove microtube-dependent tumour cell invasion, proliferation, interconnection, and radioresistance. Oligodendroglial brain tumours were deficient in this mechanism. In summary, astrocytomas can develop functional multicellular network structures. Disconnection of astrocytoma cells by targeting their tumour microtubes emerges as a new principle to reduce the treatment resistance of this disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            FoxM1 promotes β-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis.

            Wnt/β-catenin signaling is essential for stem cell regulation and tumorigenesis, but its molecular mechanisms are not fully understood. Here, we report that FoxM1 is a downstream component of Wnt signaling and is critical for β-catenin transcriptional function in tumor cells. Wnt3a increases the level and nuclear translocation of FoxM1, which binds directly to β-catenin and enhances β-catenin nuclear localization and transcriptional activity. Genetic deletion of FoxM1 in immortalized neural stem cells abolishes β-catenin nuclear localization. FoxM1 mutations that disrupt the FoxM1-β-catenin interaction or FoxM1 nuclear import prevent β-catenin nuclear accumulation in tumor cells. FoxM1-β-catenin interaction controls Wnt target gene expression, is required for glioma formation, and represents a mechanism for canonical Wnt signaling during tumorigenesis. Copyright © 2011 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Heterogeneity maintenance in glioblastoma: a social network.

              Glioblastoma multiforme (GBM), the most common intracranial tumor in adults, is characterized by extensive heterogeneity at the cellular and molecular levels. This insidious feature arises inevitably in almost all cancers and has great significance for the general outcome of the malignancy, because it confounds our understanding of the disease and also intrinsically contributes to the tumor's aggressiveness and poses an obstacle to the design of effective therapies. The classic view that heterogeneity arises as the result of a tumor's "genetic chaos" and the more contemporary cancer stem cell (CSC) hypothesis tend to identify a single cell population as the therapeutic target: the prevailing clone over time in the first case and the CSC in the latter. However, there is growing evidence that the different tumor cell populations may not be simple bystanders. Rather, they can establish a complex network of interactions between each other and with the tumor microenvironment that eventually strengthens tumor growth and increases chances to escape therapy. These differing but complementary ideas about the origin and maintenance of tumor heterogeneity and its importance in GBM are reviewed here.
                Bookmark

                Author and article information

                Contributors
                Journal
                Neurooncol Adv
                Neurooncol Adv
                noa
                Neuro-Oncology Advances
                Oxford University Press (US )
                2632-2498
                May-Dec 2019
                05 November 2019
                05 November 2019
                : 1
                : 1
                : vdz042
                Affiliations
                [1 ] Department of Obstetrics and Gynecology, University of Texas Health San Antonio , San Antonio, Texas
                [2 ] Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio , San Antonio, Texas
                [3 ] Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio , San Antonio, Texas
                [4 ] Hematology & Oncology, University of Texas Health San Antonio , San Antonio, Texas
                [5 ] Mays Cancer Center, University of Texas Health San Antonio , San Antonio, Texas
                [6 ] Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University , Augusta, Georgia
                Author notes
                Correspondence Author: Ratna K. Vadlamudi, Department of Obstetrics and Gynecology, 7703 Floyd Curl Drive, University of Texas Health San Antonio, San Antonio, TX 78229 ( vadlamudi@ 123456uthscsa.edu ).
                Article
                vdz042
                10.1093/noajnl/vdz042
                7147719
                32309805
                3dd8a0c8-3078-42d1-a3a8-2eaecf462324
                © The Author(s) 2019. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 28 November 2019
                Page count
                Pages: 14
                Funding
                Funded by: National Institutes of Health, DOI 10.13039/100000002;
                Award ID: CA178499
                Funded by: Mays Cancer Center;
                Award ID: P30CA054174-17
                Funded by: Voelcker Fund Young Investigator;
                Award ID: P30CA054174-17
                Categories
                Basic and Translational Investigations

                β-catenin signaling,coregulator,glioblastoma,glioma,pelp1
                β-catenin signaling, coregulator, glioblastoma, glioma, pelp1

                Comments

                Comment on this article