KSR1 and EPHB4 regulate Myc and PGC1β to promote survival of human colon tumors

Jamie L. McCall, Drew Gehring, Beth K. Clymer, Kurt W. Fisher, Binita Das, David L. Kelly, Hyunseok Kim, Michael A. White, Robert E. Lewis

Research output: Contribution to journalArticlepeer-review

24 Scopus citations

Abstract

Identification and characterization of survival pathways active in tumor cells but absent in normal tissues provide opportunities to develop effective anticancer therapies with reduced toxicity to the patient. We show here that, like kinase suppressor of Ras 1 (KSR1), EPH (erythropoietin-producing hepatocellular carcinoma) receptor B4 (EPHB4) is aberrantly overexpressed in human colon tumor cell lines and selectively required for their survival. KSR1 and EPHB4 support tumor cell survival by promoting the expression of downstream targets, Myc and the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1β (PGC1β). While KSR1 promotes the aberrant expression of Myc and the PGC1β protein via a posttranscriptional mechanism, EPHB4 has a greater effect on Myc and PGC1β expression via its ability to elevate mRNA levels. Subsequent analysis of the posttranscriptional regulation demonstrated that KSR1 promotes the translation of Myc protein. These findings reveal novel KSR1- and EPHB4-dependent signaling pathways supporting the survival of colorectal cancer cells through regulation of Myc and PGC1β, suggesting that inhibition of KSR1 or EPHB4 effectors may lead to selective toxicity in colorectal tumors.

Original languageEnglish (US)
Pages (from-to)2246-2261
Number of pages16
JournalMolecular and cellular biology
Volume36
Issue number17
DOIs
StatePublished - 2016

ASJC Scopus subject areas

  • Molecular Biology
  • Cell Biology

Fingerprint

Dive into the research topics of 'KSR1 and EPHB4 regulate Myc and PGC1β to promote survival of human colon tumors'. Together they form a unique fingerprint.

Cite this