Elevated FOXG1 and SOX2 in glioblastoma enforces neural stem cell identity through transcriptional control of cell cycle and epigenetic regulators

  1. Steven M. Pollard1,2
  1. 1Medical Research Council (MRC) Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom;
  2. 2Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom;
  3. 3Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom;
  4. 4Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6BT, United Kingdom;
  5. 5Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom;
  6. 6MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom;
  7. 7Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
  1. Corresponding author: steven.pollard{at}ed.ac.uk

Abstract

Glioblastoma multiforme (GBM) is an aggressive brain tumor driven by cells with hallmarks of neural stem (NS) cells. GBM stem cells frequently express high levels of the transcription factors FOXG1 and SOX2. Here we show that increased expression of these factors restricts astrocyte differentiation and can trigger dedifferentiation to a proliferative NS cell state. Transcriptional targets include cell cycle and epigenetic regulators (e.g., Foxo3, Plk1, Mycn, Dnmt1, Dnmt3b, and Tet3). Foxo3 is a critical repressed downstream effector that is controlled via a conserved FOXG1/SOX2-bound cis-regulatory element. Foxo3 loss, combined with exposure to the DNA methylation inhibitor 5-azacytidine, enforces astrocyte dedifferentiation. DNA methylation profiling in differentiating astrocytes identifies changes at multiple polycomb targets, including the promoter of Foxo3. In patient-derived GBM stem cells, CRISPR/Cas9 deletion of FOXG1 does not impact proliferation in vitro; however, upon transplantation in vivo, FOXG1-null cells display increased astrocyte differentiation and up-regulate FOXO3. In contrast, SOX2 ablation attenuates proliferation, and mutant cells cannot be expanded in vitro. Thus, FOXG1 and SOX2 operate in complementary but distinct roles to fuel unconstrained self-renewal in GBM stem cells via transcriptional control of core cell cycle and epigenetic regulators.

Keywords

Footnotes

  • Supplemental material is available for this article.

  • Article published online ahead of print. Article and publication date are online at http://www.genesdev.org/cgi/doi/10.1101/gad.293027.116.

  • Freely available online through the Genes & Development Open Access option.

  • Received November 1, 2016.
  • Accepted March 30, 2017.

This article, published in Genes & Development, is available under a Creative Commons License (Attribution 4.0 International), as described at http://creativecommons.org/licenses/by/4.0/.

| Table of Contents
OPEN ACCESS ARTICLE

Life Science Alliance