4.8 Article

Oncogenic RAS-induced CK1α drives nuclear FOXO proteolysis

Journal

ONCOGENE
Volume 37, Issue 3, Pages 363-376

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/onc.2017.334

Keywords

-

Funding

  1. Singapore Ministry of Health's National Medical Research Council under its NMRC-CBRG New Investigator Grant [NMRC/BNIG/1078/2012]
  2. Duke-NUS-St. Baldrick's Foundation Pediatric Cancer Research Fund [Duke-NUSSBF/2015/0004]
  3. Singapore Ministry of Education Academic Research Fund (AcRF) Tier 2 grant [MOE2016-T2-2-052]
  4. Singapore Agency for Science, Technology and Research (A*STAR)
  5. Singapore Ministry of Health (MOH) under its Duke-NUS Signature Research Program
  6. National Research Foundation under its Singapore Translational Research (STaR) Investigator Award
  7. Singapore Ministry of Health's National Medical Research Council [NMRC/STaR/0017/2013]

Ask authors/readers for more resources

Evasion of forkhead box O (FOXO) family of longevity-related transcription factors-mediated growth suppression is necessary to promote cancer development. Since somatic alterations or mutations and transcriptional dysregulation of the FOXO genes are infrequent in human cancers, it remains unclear how these tumour suppressors are eliminated from cancer cells. The protein stability of FOXO3A is regulated by Casein Kinase 1 alpha (CK1 alpha) in an oncogenic RAS-specific manner, but whether this mode of regulation extends to related FOXO family members is unknown. Here we report that CK1 alpha similarly destabilizes FOXO4 in RASmutant cells by phosphorylation at serines 265/268. The CK1 alpha-dependent phosphoregulation of FOXO4 is primed, in part, by the PI3K/AKT effector axis of oncogenic RAS signalling. In addition, mutant RAS coordinately elevates proteasome subunit expression and proteolytic activity to eradicate nuclear FOXO4 proteins from RAS-mutant cancer cells. Importantly, dual inhibition of CK1 alpha and the proteasome synergistically inhibited the growth of multiple RAS-mutant human cancer cell lines of diverse tissue origin by blockade of nuclear FOXO4 degradation and induction of caspase-dependent apoptosis. Our findings challenge the current paradigm that nuclear export regulates the proteolysis of FOXO3A/4 tumour suppressors in the context of cancer and illustrates how oncogenic RAS-mediated degradation of FOXOs, via post-translational mechanisms, blocks these important tumour suppressors.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available