4.7 Article

Tyrosine phosphorylation of RAS by ABL allosterically enhances effector binding

Journal

FASEB JOURNAL
Volume 29, Issue 9, Pages 3750-3761

Publisher

FEDERATION AMER SOC EXP BIOL
DOI: 10.1096/fj.15-271510

Keywords

GTPase; signal transduction; RIN1; RAF1

Funding

  1. U.S. National Institutes of Health (NIH) National Cancer Institute (NCI) Grant [CA136699]
  2. Ruth L. Kirschstein National Research Service Award Grant [GM007185]
  3. Whitcome Training Fellowship
  4. U.S. National Science Foundation Grant [MCB-1244203]
  5. NIH NCI Grant [CA168585]
  6. American Cancer Society Research Scholar Award [RSG-12-257-01-TBE]
  7. U.S. National Center for Advancing Translational Sciences Grant [UL1TR000124]
  8. University of California, Los Angeles (UCLA) Jonsson Comprehensive Cancer Center
  9. Div Of Molecular and Cellular Bioscience
  10. Direct For Biological Sciences [1244203] Funding Source: National Science Foundation

Ask authors/readers for more resources

RAS proteins are signal transduction gatekeepers that mediate cell growth, survival, and differentiation through interactions with multiple effector proteins. The RAS effector RAS- and RAB-interacting protein 1 (RIN1) activates its own downstream effectors, the small GTPase RAB5 and the tyrosine kinase Abelson tyrosine-protein kinase (ABL), to modulate endocytosis and cytoskeleton remodeling. To identify ABL substrates downstream of RAS-to-RIN1 signaling, we examined human HEK293T cells over-expressing components of this pathway. Proteomic analysis revealed several novel phosphotyrosine peptides, including Harvey rat sarcoma oncogene (HRAS)-pTyr(137). Here we report that ABL phosphorylates tyrosine 137 of H-, K-, and NRAS. Increased RIN1 levels enhanced HRAS-Tyr(137) phosphorylation by nearly 5-fold, suggesting that RAS-stimulated RIN1 can drive ABL-mediated RAS modification in a feedback circuit. Tyr(137) is well conserved among RAS orthologs and is part of a transprotein H-bond network. Crystal structures of HRAS(Y137F) and HRAS(Y137E) revealed conformation changes radiating from the mutated residue. Although consistent with Tyr(137) participation in allosteric control of HRAS function, the mutations did not alter intrinsic GTP hydrolysis rates in vitro. HRAS-Tyr(137) phosphorylation enhanced HRAS signaling capacity in cells, however, as reflected by a 4-fold increase in the association of phosphorylated HRAS(G12V) with its effector protein RAF proto-oncogene serine/threonine protein kinase 1 (RAF1). These data suggest that RAS phosphorylation at Tyr(137) allosterically alters protein conformation and effector binding, providing a mechanism for effector-initiated modulation of RAS signaling.

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