4.7 Article

C3G downregulation induces the acquisition of a mesenchymal phenotype that enhances aggressiveness of glioblastoma cells

Journal

CELL DEATH & DISEASE
Volume 12, Issue 4, Pages -

Publisher

SPRINGERNATURE
DOI: 10.1038/s41419-021-03631-w

Keywords

-

Categories

Funding

  1. Spanish Ministry of Economy and Competitiveness [SAF2016-76588-C2-1-R, PID2019-104143RB-C22, SAF2016-76588-C2-2-R, PID2019-104143RB-C21]
  2. Council of Education of Junta de Castilla y Leon, Spain [SA017U16, SA078P20]
  3. European FEDER Program
  4. Spanish Ministry of Education
  5. Complutense University from Madrid
  6. Madrid Community Program for Talent Attraction [MRF 2017-T1/BMD-5468]
  7. BBVA (Becas Leonardo 2018) [BBM-TRA-0041]

Ask authors/readers for more resources

The downregulation of C3G in GBM promotes migration and invasion of GBM cells, affects proliferation, impairs EGFR signaling, and upregulates activation of other receptor tyrosine kinases through different pathways. The levels of C3G may be crucial in discriminating GBM patient responders to various RTK inhibition protocols. Patients with low C3G expression may not respond well to EGFR inhibitors.
Glioblastoma (GBM) is the most aggressive tumor from the central nervous system (CNS). The current lack of efficient therapies makes essential to find new treatment strategies. C3G, a guanine nucleotide exchange factor for some Ras proteins, plays a dual role in cancer, but its function in GBM remains unknown. Database analyses revealed a reduced C3G mRNA expression in GBM patient samples. C3G protein levels were also decreased in a panel of human GBM cell lines as compared to astrocytes. Based on this, we characterized C3G function in GBM using in vitro and in vivo human GBM models. We report here that C3G downregulation promoted the acquisition of a more mesenchymal phenotype that enhanced the migratory and invasive capacity of GBM cells. This facilitates foci formation in anchorage-dependent and -independent growth assays and the generation of larger tumors in xenografts and chick chorioallantoic membrane (CAM) assays, but with a lower cell density, as proliferation was reduced. Mechanistically, C3G knock-down impairs EGFR signaling by reducing cell surface EGFR through recycling inhibition, while upregulating the activation of several other receptor tyrosine kinases (RTKs) that might promote invasion. In particular, FGF2, likely acting through FGFR1, promoted invasion of C3G-silenced GBM cells. Moreover, ERKs mediate this invasiveness, both in response to FGF2- and serum-induced chemoattraction. In conclusion, our data show the distinct dependency of GBM tumors on C3G for EGF/EGFR signaling versus other RTKs, suggesting that assessing C3G levels may discriminate GBM patient responders to different RTK inhibition protocols. Hence, patients with a low C3G expression might not respond to EGFR inhibitors.

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