SOX2-RNAi attenuates S-phase entry and induces RhoA-dependent switch to protease-independent amoeboid migration in human glioma cells

Research output: Contribution to journalResearch articleContributedpeer-review

Contributors

  • Felix Oppel - , University Hospital Carl Gustav Carus Dresden, Chirurgische Intensivstation (ZCH-ITS) (Author)
  • Nadja Müller - , University Hospital Carl Gustav Carus Dresden, Department of Neurosurgery (Author)
  • Gabriele Schackert - , University Hospital Carl Gustav Carus Dresden, Department of Neurosurgery (Author)
  • Sandy Hendruschk - , University Hospital Carl Gustav Carus Dresden, Department of Neurosurgery (Author)
  • Daniel Martin - , Department of Neurosurgery, University Hospital Carl Gustav Carus Dresden (Author)
  • Kathrin D. Geiger - , Institute of Pathology (Author)
  • Achim Temme - , Department of Neurosurgery, University Hospital Carl Gustav Carus Dresden (Author)

Abstract

Background: SOX2, a high mobility group (HMG)-box containing transcription factor, is a key regulator during development of the nervous system and a persistent marker of neural stem cells. Recent studies suggested a role of SOX2 in tumor progression. In our previous work we detected SOX2 in glioma cells and glioblastoma specimens. Herein, we aim to explore the role of SOX2 for glioma malignancy in particular its role in cell proliferation and migration.Methods: Retroviral shRNA-vectors were utilized to stably knockdown SOX2 in U343-MG and U373-MG cells. The resulting phenotype was investigated by Western blot, migration/invasion assays, RhoA G-LISA, time lapse video imaging, and orthotopic xenograft experiments.Results: SOX2 depletion results in pleiotropic effects including attenuated cell proliferation caused by decreased levels of cyclinD1. Also an increased TCF/LEF-signaling and concomitant decrease in Oct4 and Nestin expression was noted. Furthermore, down-regulation of focal adhesion kinase (FAK) signaling and of downstream proteins such as HEF1/NEDD9, matrix metalloproteinases pro-MMP-1 and -2 impaired invasive proteolysis-dependent migration. Yet, cells with knockdown of SOX2 switched to a RhoA-dependent amoeboid-like migration mode which could be blocked by the ROCK inhibitor Y27632 downstream of RhoA-signaling. Orthotopic xenograft experiments revealed a higher tumorigenicity of U343-MG glioma cells transduced with shRNA targeting SOX2 which was characterized by increased dissemination of glioma cells.Conclusion: Our findings suggest that SOX2 plays a role in the maintenance of a less differentiated glioma cell phenotype. In addition, the results indicate a critical role of SOX2 in adhesion and migration of malignant gliomas.

Details

Original languageEnglish
Article number137
JournalMolecular cancer
Volume10
Publication statusPublished - 9 Nov 2011
Peer-reviewedYes

External IDs

PubMed 22070920

Keywords

Sustainable Development Goals