4.7 Article

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

Journal

GENES & DEVELOPMENT
Volume 31, Issue 8, Pages 757-773

Publisher

COLD SPRING HARBOR LAB PRESS, PUBLICATIONS DEPT
DOI: 10.1101/gad.293027.116

Keywords

glioblastoma; cell cycle; epigenetics; dedifferentiation; neural stem cell; astrocyte

Funding

  1. Wellcome Trust Clinician Research Training Fellowship
  2. Biotechnology and Biological Sciences Research Council
  3. EMBO
  4. Cancer Research UK [A19680]
  5. Science Without Borders Program (CAPES, Brazil)
  6. Medical Research Council University
  7. Biotechnology and Biological Sciences Research Council [BB/M018040/1] Funding Source: researchfish
  8. Cancer Research UK [16399, 17368, 20837] Funding Source: researchfish
  9. Medical Research Council [MC_PC_12009, MR/K017047/1, MR/J013137/1] Funding Source: researchfish
  10. The Brain Tumour Charity [8/105, GN-000358] Funding Source: researchfish
  11. Versus Arthritis
  12. Cancer Research UK [21992] Funding Source: researchfish
  13. BBSRC [BB/M018040/1] Funding Source: UKRI
  14. MRC [MR/K017047/1, MR/J013137/1] Funding Source: UKRI

Ask authors/readers for more resources

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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available