4.6 Article

Identification of Radil as a Ras binding partner and putative activator

Journal

JOURNAL OF BIOLOGICAL CHEMISTRY
Volume 296, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.jbc.2021.100314

Keywords

-

Funding

  1. US Public Service Awards [CA213159, CA216987]
  2. NIEHS Center grant [ES000260]

Ask authors/readers for more resources

Radil, a downstream effector of Ras, interacts strongly with KRas and GTP-bound Ras, suggesting a role in Ras activation. Overexpression of Radil leads to transient activation of MAPK signaling, while Radil knockdown weakens downstream Ras signaling, affecting cell proliferation, invasion, and migration. Radil plays a vital role in regulating Ras signaling, cell adhesion, and the epithelial-mesenchymal transition, potentially offering new opportunities for Ras-related anticancer drug development.
Ras genes are among the most frequently mutated oncogenes in human malignancies. To date, there are no successful anticancer drugs in the clinic that target Ras proteins or their pathways. Therefore, it is imperative to identify and characterize new components that regulate Ras activity or mediate its downstream signaling. To this end, we used a combination of affinity-pulldown and mass spectrometry to search for proteins that are physically associated with KRas. One of the top hits was Radil, a gene product with a Ras-association domain. Radil is known to be a downstream effector of Rap1, inhibiting RhoA signaling to regulate cell adhesion and migration. We demonstrate that Radil interacted with all three isoforms of Ras including HRas, NRas, and KRas, although it exhibited the strongest interaction with KRas. Moreover, Radil interacts with GTP-bound Ras more efficiently, suggesting a possibility that Radil may be involved in Ras activation. Supporting this, ectopic expression of Radil led to transient activation of mitogen-activated protein kinase kinase and extracellular signal-regulated kinase; Radil knockdown resulted in weakened activation of Ras downstream signaling components, which was coupled with decreased cell proliferation and invasion, and reduced expression of mesenchymal cell markers. Moreover, Radil knockdown greatly reduced the number of adhesion foci and depolymerized actin filaments, molecular processes that facilitate cancer cell migration. Taken together, our present studies strongly suggest that Radil is an important player for regulating Ras signaling, cell adhesion, and the epithelial-mesenchymal transition and may provide new directions for Ras-related anticancer drug development.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available