RAS mutations affect tumor necrosis factor-induced apoptosis in colon carcinoma cells via ERK-modulatory negative and positive feedback circuits along with non-ERK pathway effects.

TitleRAS mutations affect tumor necrosis factor-induced apoptosis in colon carcinoma cells via ERK-modulatory negative and positive feedback circuits along with non-ERK pathway effects.
Publication TypeJournal Article
Year of Publication2009
AuthorsKreeger, PK, Mandhana, R, Alford, SK, Haigis, KM, Lauffenburger, DA
JournalCancer Res
Volume69
Issue20
Pagination8191-9
Date Published2009 Oct 15
ISSN1538-7445
KeywordsApoptosis, Autocrine Communication, Blotting, Western, Chemokines, Colonic Neoplasms, Computational Biology, Dual-Specificity Phosphatases, Enzyme-Linked Immunosorbent Assay, Extracellular Signal-Regulated MAP Kinases, Feedback, Physiological, Flow Cytometry, Fluorescent Antibody Technique, Genes, ras, Humans, Mutation, Proto-Oncogene Proteins, ras Proteins, Reverse Transcriptase Polymerase Chain Reaction, RNA, Messenger, Transforming Growth Factor beta, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha
Abstract

More than 40% of colon cancers have a mutation in K-RAS or N-RAS, GTPases that operate as central hubs for multiple key signaling pathways within the cell. Utilizing an isogenic panel of colon carcinoma cells with K-RAS or N-RAS variations, we observed differences in tumor necrosis factor-alpha (TNFalpha)-induced apoptosis. When the dynamics of phosphorylated ERK response to TNFalpha were examined, K-RAS mutant cells showed lower activation whereas N-RAS mutant cells exhibited prolonged duration. These divergent trends were partially explained by differential induction of two ERK-modulatory circuits: negative feedback mediated by dual-specificity phosphatase 5 and positive feedback by autocrine transforming growth factor-alpha. Moreover, in the various RAS mutant colon carcinoma lines, the transforming growth factor-alpha autocrine loop differentially elicited a further downstream chemokine (CXCL1/CXCL8) autocrine loop, with the two loops having opposite effects on apoptosis. Although the apoptotic responses of the RAS mutant panel to TNFalpha treatment showed significant dependence on the respective phosphorylated ERK dynamics, successful prediction across the various cell lines required contextual information concerning additional pathways including IKK and p38. A quantitative computational model based on weighted linear combinations of these pathway activities successfully predicted not only the spectrum of cell death responses but also the corresponding chemokine production responses. Our findings indicate that diverse RAS mutations yield differential cell behavioral responses to inflammatory cytokine exposure by means of (a) differential effects on ERK activity via multiple feedback circuit mechanisms, and (b) differential effects on other key signaling pathways contextually modulating ERK-related dependence.

DOI10.1158/0008-5472.CAN-09-1921
Alternate JournalCancer Res.
PubMed ID19789336
PubMed Central IDPMC2927135
Grant ListP50-GM68762 / GM / NIGMS NIH HHS / United States
U54 CA112967-05 / CA / NCI NIH HHS / United States
U54-CA112967 / CA / NCI NIH HHS / United States