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.

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.

[1]  B. LaFleur,et al.  TACE/ADAM-17: A Component of the Epidermal Growth Factor Receptor Axis and a Promising Therapeutic Target in Colorectal Cancer , 2008, Clinical Cancer Research.

[2]  H. Wiley,et al.  Multiple Mechanisms Are Responsible for Transactivation of the Epidermal Growth Factor Receptor in Mammary Epithelial Cells* , 2008, Journal of Biological Chemistry.

[3]  Douglas A. Lauffenburger,et al.  Common effector processing mediates cell-specific responses to stimuli , 2007, Nature.

[4]  Aihua Li,et al.  Constitutive expression of growth regulated oncogene (gro) in human colon carcinoma cells with different metastatic potential and its role in regulating their metastatic phenotype , 2005, Clinical & Experimental Metastasis.

[5]  S. Christmas,et al.  Interleukin-8 as an autocrine growth factor for human colon carcinoma cells in vitro. , 2000, Cytokine.

[6]  A. Wolfman,et al.  Endogenous c-N-Ras Provides a Steady-state Anti-apoptotic Signal* , 2000, The Journal of Biological Chemistry.

[7]  L. Augenlicht,et al.  Oncogenic Ras Promotes Butyrate-induced Apoptosis through Inhibition of Gelsolin Expression* , 2004, Journal of Biological Chemistry.

[8]  Jonathan Melamed,et al.  Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence , 2008, Cell.

[9]  John G. Albeck,et al.  Cue-Signal-Response Analysis of TNF-Induced Apoptosis by Partial Least Squares Regression of Dynamic Multivariate Data , 2004, J. Comput. Biol..

[10]  D. Hommes,et al.  New cytokine therapeutics for inflammatory bowel disease. , 2004, Cytokine.

[11]  B. Kowalski,et al.  Partial least-squares regression: a tutorial , 1986 .

[12]  R. Coffey,et al.  Oncogenic KRAS provides a uniquely powerful and variable oncogenic contribution among RAS family members in the colonic epithelium , 2007, Journal of cellular physiology.

[13]  L. Samson,et al.  DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. , 2008, The Journal of clinical investigation.

[14]  T. Jacks,et al.  Oncogenic K-RAS subverts the antiapoptotic role of N-RAS and alters modulation of the N-RAS: gelsolin complex , 2007, Oncogene.

[15]  D. Lauffenburger,et al.  An inducible autocrine cascade regulates rat hepatocyte proliferation and apoptosis responses to tumor necrosis factor‐α , 2008, Hepatology.

[16]  T. Muto,et al.  Genetic Alterations in Ulcerative Colitis‐associated Neoplasia Focusing on APC, K‐ras Gene and Microsatellite Instability , 1999, Japanese journal of cancer research : Gann.

[17]  S. Shirasawa,et al.  Altered growth of human colon cancer cell lines disrupted at activated Ki-ras. , 1993, Science.

[18]  Melissa L. Kemp,et al.  Quantitative Network Signal Combinations Downstream of TCR Activation Can Predict IL-2 Production Response1 , 2007, The Journal of Immunology.

[19]  Eytan Domany,et al.  A module of negative feedback regulators defines growth factor signaling , 2007, Nature Genetics.

[20]  Alan Wells,et al.  EGF-receptor-mediated mammary epithelial cell migration is driven by sustained ERK signaling from autocrine stimulation , 2007, Journal of Cell Science.

[21]  D. Lauffenburger,et al.  A Compendium of Signals and Responses Triggered by Prodeath and Prosurvival Cytokines*S , 2005, Molecular & Cellular Proteomics.

[22]  S. Keyse,et al.  Differential regulation of MAP kinase signalling by dual-specificity protein phosphatases , 2007, Oncogene.

[23]  Y. Nakamura,et al.  Genetic alterations during colorectal-tumor development. , 1988, The New England journal of medicine.

[24]  J. Downward Targeting RAS signalling pathways in cancer therapy , 2003, Nature Reviews Cancer.

[25]  D. Lauffenburger,et al.  The Response of Human Epithelial Cells to TNF Involves an Inducible Autocrine Cascade , 2006, Cell.

[26]  D. Hanahan,et al.  The Hallmarks of Cancer , 2000, Cell.

[27]  S. Soond,et al.  ERK-mediated phosphorylation of Thr735 in TNFα-converting enzyme and its potential role in TACE protein trafficking , 2005, Journal of Cell Science.

[28]  C. Der,et al.  Cellular N-Ras Promotes Cell Survival by Downregulation of Jun N-Terminal Protein Kinase and p38 , 2002, Molecular and Cellular Biology.

[29]  S. Aaronson,et al.  A novel dual specificity phosphatase induced by serum stimulation and heat shock. , 1994, The Journal of biological chemistry.

[30]  M. Barbacid,et al.  RAS oncogenes: the first 30 years , 2003, Nature Reviews Cancer.

[31]  Martin Vingron,et al.  A systems biological approach suggests that transcriptional feedback regulation by dual‐specificity phosphatase 6 shapes extracellular signal‐related kinase activity in RAS‐transformed fibroblasts , 2009, The FEBS journal.

[32]  D. Lauffenburger,et al.  A Systems Model of Signaling Identifies a Molecular Basis Set for Cytokine-Induced Apoptosis , 2005, Science.

[33]  B. Vogelstein,et al.  Prevalence of ras gene mutations in human colorectal cancers , 1987, Nature.

[34]  Pietro Ghezzi,et al.  Noncompetitive allosteric inhibitors of the inflammatory chemokine receptors CXCR1 and CXCR2: prevention of reperfusion injury. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[35]  Michael B. Yaffe,et al.  Data-driven modelling of signal-transduction networks , 2006, Nature Reviews Molecular Cell Biology.

[36]  J. Mayberry,et al.  Colorectal cancer complicating ulcerative colitis: a review , 2000, American Journal of Gastroenterology.

[37]  G. Kollias,et al.  Impaired on/off regulation of TNF biosynthesis in mice lacking TNF AU-rich elements: implications for joint and gut-associated immunopathologies. , 1999, Immunity.

[38]  Forest M. White,et al.  Modeling HER2 Effects on Cell Behavior from Mass Spectrometry Phosphotyrosine Data , 2006, PLoS Comput. Biol..

[39]  D. Notterman,et al.  GROα Is Highly Expressed in Adenocarcinoma of the Colon and Down-Regulates Fibulin-1 , 2006, Clinical Cancer Research.

[40]  A. Sweet-Cordero,et al.  Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon , 2008, Nature Genetics.