9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CHL1 Is Expressed and Functions as a Malignancy Promoter in Glioma 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

          The cell adhesion molecule with homology to L1CAM (close homolog of L1) (CHL1) is a member of the cell adhesion molecule L1 (L1CAM) gene family. Although CHL1 expression and function have been reported in several tumors, the roles of CHL1 in the development of glioma remain unclear. In the present study, we investigated the effects of CHL1 on proliferation indexes and activation of Akt1 and Erk signaling by siRNA in U-87 MG human glioblastoma and human U251 and SHG-44 glioma cells. We found that siRNA targeting CHL1 significantly down-regulated the expression of CHL1 mRNA and protein accompanied by reduced cell proliferation and transmigration invasion in all three cell lines. Down-regulating CHL1 expression also reduced cell survival, as measured by the Bax/Bcl-2 ratio, and increased activation of caspase-3. In subcutaneous U-87 MG cell xenograft tumors in nude mice, intratumoral administration of siRNA targeting CHL1 treatment significantly down-regulated CHL1 expression in vivo, accompanied by increased levels of activated caspase-3. Our combined results confirmed for the first time that in contrast to findings about CHL1 in most other cancer types, CHL1 functions in promoting cell proliferation, metastasis and migration in human glioma cells both in vitro and in vivo. These results indicate that CHL1 is a therapeutic target in the clinical management of glioma/glioblastoma.

          Related collections

          Most cited references45

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

          Senescence in tumours: evidence from mice and humans.

          The importance of cellular senescence, which is a stress response that stably blocks proliferation, is increasingly being recognized. Senescence is prevalent in pre-malignant tumours, and progression to malignancy requires evading senescence. Malignant tumours, however, may still undergo senescence owing to interventions that restore tumour suppressor function or inactivate oncogenes. Senescent tumour cells can be cleared by immune cells, which may result in efficient tumour regression. Standard chemotherapy also has the potential to induce senescence, which may partly underlie its therapeutic activity. Although these concepts are well supported in mouse models, translating them to clinical oncology remains a challenge.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Elevated invasive potential of glioblastoma stem cells.

            Glioblastomas (GBMs) are the most lethal and common types of primary brain tumors. The hallmark of GBMs is their highly infiltrative nature. The cellular and molecular mechanisms underlying the aggressive cancer invasion in GBMs are poorly understood. GBM displays remarkable cellular heterogeneity and hierarchy containing self-renewing glioblastoma stem cells (GSCs). Whether GSCs are more invasive than non-stem tumor cells and contribute to the invasive phenotype in GBMs has not been determined. Here we provide experimental evidence supporting that GSCs derived from GBM surgical specimens or xenografts display greater invasive potential in vitro and in vivo than matched non-stem tumor cells. Furthermore, we identified several invasion-associated proteins that were differentially expressed in GSCs relative to non-stem tumor cells. One of such proteins is L1CAM, a cell surface molecule shown to be critical to maintain GSC tumorigenic potential in our previous study. Immunohistochemical staining showed that L1CAM is highly expressed in a population of cancer cells in the invasive fronts of primary GBMs. Collectively, these data demonstrate the invasive nature of GSCs, suggesting that disrupting GSCs through a specific target such as L1CAM may reduce GBM cancer invasion and tumor recurrence. Copyright © 2011 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              MicroRNA-10a targets CHL1 and promotes cell growth, migration and invasion in human cervical cancer cells.

              MicroRNAs (miRNAs) play an important role in cancer initiation, progression and metastasis by regulating their target genes. Here, we found microRNA-10a (miR-10a) is upregulated in human cervical cancer and promotes the colony formation activity, migration and invasion of HeLa and C33A cells. Subsequently, CHL1 is confirmed as a target of miR-10a and is negatively regulated by miR-10a at mRNA and protein levels. Furthermore, knockdown of CHL1 expression results in increased colony formation activity, migration and invasion. Finally, overexpression of CHL1 lacked the 3'UTR abolished the effects of miR-10a. Our results may provide a strategy for blocking tumor metastasis. Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Mol Neurosci
                Front Mol Neurosci
                Front. Mol. Neurosci.
                Frontiers in Molecular Neuroscience
                Frontiers Media S.A.
                1662-5099
                17 October 2017
                2017
                : 10
                : 324
                Affiliations
                [1] 1Center for Neuroscience, Shantou University Medical College , Shantou, China
                [2] 2Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University , Piscataway, NJ, United States
                Author notes

                Edited by: Vladimir Parpura, University of Alabama at Birmingham, United States

                Reviewed by: Marcus Semtner, Max Delbrück Center for Molecular Medicine (HZ), Germany; Yonghe Wu, Deutsches Krebsforschungszentrum (DKFZ), Germany

                *Correspondence: Melitta Schachner schachner@ 123456stu.edu.cn Wei-Jiang Zhao neuromancn@ 123456aliyun.com

                These authors have contributed equally to this work.

                Article
                10.3389/fnmol.2017.00324
                5650976
                29089868
                aa48ff55-75bd-4dfa-837e-2f8d279328bb
                Copyright © 2017 Yang, Xie, Hu, Jiang, Shen, Schachner and Zhao.

                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) or licensor 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
                : 13 June 2017
                : 25 September 2017
                Page count
                Figures: 10, Tables: 1, Equations: 0, References: 51, Pages: 14, Words: 8617
                Funding
                Funded by: National Natural Science Foundation of China 10.13039/501100001809
                Award ID: 81471279, 81171138
                Categories
                Neuroscience
                Original Research

                Neurosciences
                glioma,glioma cell lines,close homolog of l1 cell adhesion molecule (chl1),sirna,malignancy

                Comments

                Comment on this article