Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-vfjqv Total loading time: 0 Render date: 2024-04-26T11:54:48.122Z Has data issue: false hasContentIssue false

21 - Ras

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Adrienne D. Cox
Affiliation:
Departments of Radiation Oncology and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
Molly J. DeCristo
Affiliation:
Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Ras

First identified in transforming retroviruses, Ras proteins are GTP/GDP-binding molecular switches and the founding members of the Ras superfamily of small GTPases (1,2). They serve as key signaling nodes mediating many aspects of normal cell physiology, such as mitogen-stimulated growth, differentiation and death/survival, and are the most frequently mutated oncogenes in human cancers. The activity of Ras proteins is regulated by a cycle of binding to guanine nucleotides, where the GDP-bound protein is in the resting state, and the GTP-bound protein is in the active form (Figure 21.1). Activation is induced by positive regulators called guanine nucleotide exchange factors (GEFs) that promote dissociation of the bound GDP. Due to the high intra-cellular concentrations of GTP, this dissociation results in the exchange of GDP for GTP. Once bound to GTP, Ras assumes a conformation that promotes binding to its downstream effector proteins and subsequent transmission of signals that produce its biological effects. Normal Ras signaling is then rapidly turned off by the action of negative regulatory GTPase activating proteins (GAPs) that accelerate the otherwise slow rate of intrinsic hydrolysis of the bound GTP to GDP, thereby restoring the resting GDP-bound conformation. Ras proteins that are resistant to GAP activity are constitutively GTP-bound and active. Mutational activation of RAS that causes the protein to be GAP-resistant is the most common mechanism of RAS-mediated oncogenesis. The three RAS genes, HRAS, KRAS, and NRAS, specify four ~21 kDa Ras proteins, with alternative splicing of the fourth exon of KRAS resulting in expression of K-Ras4A and K-Ras4B proteins. The Ras isoforms are not fully inter-changeable. In addition to GTP-binding, all Ras proteins require modification by farnesyl isoprenoids at their C-terminal CAAX motifs for their subcellular localization and biological activity; the four isoforms vary in the additional post-translational modifications that are signaled by their distinct C-terminal membrane-targeting domains. These distinctions play a large role in determining the specific spatiotemporal regulation and biological consequences of both wild-type and oncogenic forms of Ras. Although the particular RAS isoform and residue vary among cancer types, missense mutations in KRAS at G12 are by far the most frequent, especially in cancers of the pancreas, colon, and lung, followed by NRAS Q61 mutations, especially in hematopoietic diseases and melanomas; HRAS is rarely mutated.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 258 - 271
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Cox, AD, Der, CJ. Ras history: The saga continues. Small GTPases 2010;1:2–27.CrossRef
Malumbres, M, Barbacid, M. RAS oncogenes: the first 30 years. Nature Reviews Cancer 2003;3:459–65.CrossRef
Colicelli, J. Human RAS superfamily proteins and related GTPases. Science STKE 2004:RE13.
Wennerberg, K, Rossman, KL, Der, CJ. The Ras superfamily at a glance. Journal of Cell Science 2005;118:843–6.CrossRefGoogle ScholarPubMed
Jiang, SY, Ramachandran, S. Comparative and evolutionary analysis of genes encoding small GTPases and their activating proteins in eukaryotic genomes. Physiological Genomics 2006;24:235–51.CrossRef
Beitel, GJ, Clark, SG, Horvitz, HR. Caenorhabditis elegans ras gene let-60 acts as a switch in the pathway of vulval induction. Nature 1990;348:503–9.CrossRef
Zhang, K, DeClue, JE, Vass, WC, et al. Suppression of c-ras transformation by GTPase-activating protein. Nature 1990;346:754–6.CrossRef
Fortini, ME, Simon, MA, Rubin, GM. Signalling by the sevenless protein tyrosine kinase is mimicked by Ras1 activation. Nature 1992;355:559–61.CrossRef
Satoh, T, Endo, M, Nakafuku, M, et al. Accumulation of p21ras.GTP in response to stimulation with epidermal growth factor and oncogene products with tyrosine kinase activity. Proceedings of the National Academy of Sciences USA 1990;87:7926–9.CrossRef
Lowenstein, EJ, Daly, RJ, Batzer, AG, et al. The SH2 and SH3 domain-containing protein GRB2 links receptor tyrosine kinases to Ras signaling. Cell 1992;70:431–42.CrossRef
Olivier, JP, Raabe, T, Henkemeyer, M, et al. A Drosophila SH2-SH3 adaptor protein implicated in coupling the sevenless tyrosine kinase to an activator of Ras guanine nucleotide exchange, Sos. Cell 1993;73:179–91.CrossRef
Dickson, B, Sprenger, F, Morrison, D, Hafen, E. Raf functions downstream of Ras1 in the Sevenless signal transduction pathway. Nature 1992;360:600–3.CrossRef
Han, M, Golden, A, Han, Y, Sternberg, PW. C. elegans lin-45 raf gene participates in let-60 ras-stimulated vulval differentiation. Nature 1993;363:133–40.CrossRef
Van Aelst, L, Barr, M, Marcus, S, Polverino, A, Wigler, M. Complex formation between RAS and RAF and other protein kinases. Proceedings of the National Academy of Sciences USA 1993;90:6213–17.CrossRef
Warne, PH, Viciana, PR, Downward, J. Direct interaction of Ras and the amino-terminal region of Raf-1 in vitro. Nature 1993;364:352–5.CrossRef
Zhang, XF, Settleman, J, Kyriakis, JM, et al. Normal and oncogenic p21ras proteins bind to the amino-terminal regulatory domain of c-Raf-1. Nature 1993;364:308–13.CrossRef
Sternberg, PW, Han, M. Genetics of RAS signaling in C. elegans. Trends in Genetics 1998;14:466–72.
Tamanoi, F. Ras signaling in yeast. Genes and Cancer 2011;2:210–15.CrossRef
Haigis, KM, Kendall, KR, Wang, Y, et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nature Genetics 2008;40:600–8.CrossRef
Tuveson, DA, Shaw, AT, Willis, NA, et al. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 2004;5:375–87.CrossRef
Guerra, C, Mijimolle, N, Dhawahir, A, et al. Tumor induction by an endogenous K-ras oncogene is highly dependent on cellular context. Cancer Cell 2003;4:111–20.CrossRef
Olive, KP, Jacobetz, MA, Davidson, CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009;324:1457–61.CrossRef
Hancock, JF. Ras proteins: different signals from different locations. Nature Reviews Molecular and Cellular Biology 2003;4:373–84.CrossRef
Karnoub, AE, Weinberg, RA. Ras oncogenes: split personalities. Nature Reviews Molecular and Cellular Biology 2008;9:517–31.CrossRef
Schubbert, S, Shannon, K, Bollag, G. Hyperactive Ras in developmental disorders and cancer. Nature Reviews Cancer 2007;7:295–308.CrossRef
Yamagata, K, Sanders, LK, Kaufmann, WE, et al. rheb, a growth factor- and synaptic activity-regulated gene, encodes a novel Ras-related protein. Journal of Biological Chemistry 1994;269:16 333–9.Google ScholarPubMed
Zhang, Y, Gao, X, Saucedo, LJ, et al. Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nature Cell Biology 2003;5:578–81.CrossRef
Bodemann, BO, White, MA. Ral GTPases and cancer: linchpin support of the tumorigenic platform. Nature Reviews Cancer 2008;8:133–40.CrossRef
Wozniak, MA, Kwong, L, Chodniewicz, D, Klemke, RL, Keely, PJ. R-Ras controls membrane protrusion and cell migration through the spatial regulation of Rac and Rho. Molecular Biology of the Cell 2005;16:84–96.CrossRef
Lin, KB, Tan, P, Freeman, SA, et al. The Rap GTPases regulate the migration, invasiveness and in vivo dissemination of B-cell lymphomas. Oncogene 2010;29:608–15.CrossRef
Milburn, MV, Tong, L, deVos, AM, et al. Molecular switch for signal transduction: structural differences between active and inactive forms of protooncogenic ras proteins. Science 1990;247:939–45.CrossRef
Bos, JL, Rehmann, H, Wittinghofer, A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 2007;129:865–77.CrossRef
Feig, LA, Cooper, GM. Inhibition of NIH 3T3 cell proliferation by a mutant ras protein with preferential affinity for GDP. Molecular and Cellular Biology 1988;8:3235–43.CrossRef
Vigil, D, Cherfils, J, Rossman, KL, Der, CJ. Ras superfamily GEFs and GAPs: validated and tractable targets for cancer therapy? Nature Reviews Cancer 2010;10:842–57.
Scheffzek, K, Lautwein, A, Kabsch, W, Ahmadian, MR, Wittinghofer, A. Crystal structure of the GTPase-activating domain of human p120GAP and implications for the interaction with Ras. Nature 1996;384:591–6.CrossRef
Martin, GA, Viskochil, D, Bollag, G, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell 1990;63:843–9.CrossRef
Scheffzek, K, Ahmadian, MR, Kabsch, W, et al. The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science 1997;277:333–8.CrossRef
Goody, RS, Pai, EF, Schlichting, I, et al. Studies on the structure and mechanism of H-ras p21. Philosophical Transactions of the Royal Society London B, Biological Sciences 1992;336:3–10.CrossRef
Al-Mulla, F, Milner-White, EJ, Going, JJ, Birnie, GD. Structural differences between valine-12 and aspartate-12 Ras proteins may modify carcinoma aggression. Journal of Pathology 1999;187:433–8.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
Seeburg, PH, Colby, WW, Capon, DJ, Goeddel, DV, Levinson, AD. Biological properties of human c-Ha-ras1 genes mutated at codon 12. Nature 1984;312:71–5.CrossRef
Bos, JL. ras oncogenes in human cancer: a review. Cancer Research 1989;49:4682–9.
Shirouzu, M, Koide, H, Fujita-Yoshigaki, J, et al. Mutations that abolish the ability of Ha-Ras to associate with Raf-1. Oncogene 1994;9:2153–7.
Prior, IA, Hancock, JF. Ras trafficking, localization and compartmentalized signalling. Seminars in Cell and Developmental Biology 2012;23:145–53.CrossRef
Schafer, WR, Kim, R, Sterne, R, et al. Genetic and pharmacological suppression of oncogenic mutations in ras genes of yeast and humans. Science 1989;245:379–85.CrossRef
Casey, PJ, Solski, PA, Der, CJ, Buss, JE. p21ras is modified by a farnesyl isoprenoid. Proceedings of the National Academy of Sciences USA 1989;86:8323–7.CrossRef
Hancock, JF, Magee, AI, Childs, JE, Marshall, CJ. All ras proteins are polyisoprenylated but only some are palmitoylated. Cell 1989;57:1167–77.CrossRef
Kim, E, Ambroziak, P, Otto, JC, et al. Disruption of the mouse Rce1 gene results in defective Ras processing and mislocalization of Ras within cells. Journal of Biological Chemistry 1999;274:8383–90.CrossRefGoogle ScholarPubMed
Bergo, MO, Ambroziak, P, Gregory, C, et al. Absence of the CAAX endoprotease Rce1: effects on cell growth and transformation. Molecular and Cellular Biology 2002;22:171–81.CrossRef
Wahlstrom, AM, Cutts, BA, Karlsson, C, et al. Rce1 deficiency accelerates the development of K-RAS-induced myeloproliferative disease. Blood 2007;109:763–8.CrossRef
Bergo, MO, Gavino, BJ, Hong, C, et al. Inactivation of Icmt inhibits transformation by oncogenic K-Ras and B-Raf. Journal of Clinical Investigation 2004;113:539–50.CrossRefGoogle ScholarPubMed
Wahlstrom, AM, Cutts, BA, Liu, M, et al. Inactivating Icmt ameliorates K-RAS-induced myeloproliferative disease. Blood 2008;112:1357–65.CrossRef
Ahearn, IM, Haigis, K, Bar-Sagi, D, Philips, MR. Regulating the regulator: post-translational modification of RAS. Nature Reviews Molecular and Cellular Biology 2012;13:39–51.CrossRef
James, GL, Brown, MS, Cobb, MH, Goldstein, JL. Benzodiazepine peptidomimetic BZA-5B interrupts the MAP kinase activation pathway in H-Ras-transformed Rat-1 cells, but not in untransformed cells. Journal of Biological Chemistry 1994;269:27 705–14.Google ScholarPubMed
James, GL, Goldstein, JL, Brown, MS. Polylysine and CVIM sequences of K-RasB dictate specificity of prenylation and confer resistance to benzodiazepine peptidomimetic in vitro. Journal of Biological Chemistry 1995;270:6221–6.CrossRefGoogle ScholarPubMed
Fiordalisi, JJ, Johnson, RL, 2nd, Weinbaum CA, et al. High affinity for farnesyltransferase and alternative prenylation contribute individually to K-Ras4B resistance to farnesyltransferase inhibitors. Journal of Biological Chemistry 2003;278:41 718–27.CrossRefGoogle Scholar
Macdonald, JS, McCoy, S, Whitehead RP, et al. A Phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: a Southwest oncology group (SWOG 9924) study. Investigational New Drugs 2005;23:485–7.CrossRef
Winter-Vann, AM, Casey, PJ. Post-prenylation-processing enzymes as new targets in oncogenesis. Nature Reviews Cancer 2005;5:405–12.CrossRef
Wang, M, Tan, W, Zhou, J, et al. A small molecule inhibitor of isoprenylcysteine carboxymethyltransferase induces autophagic cell death in PC3 prostate cancer cells. Journal of Biological Chemistry 2008;283:18 678–84.CrossRefGoogle ScholarPubMed
Hancock, JF, Cadwallader, K, Paterson, H, Marshall, CJ. A CAAX or a CAAL motif and a second signal are sufficient for plasma membrane targeting of ras proteins. EMBO Journal 1991;10:4033–9.
Choy, E, Chiu, VK, Silletti, J, et al. Endomembrane trafficking of ras: the CAAX motif targets proteins to the ER and Golgi. Cell 1999;98:69–80.CrossRef
Bivona, TG, Quatela, SE, Bodemann, BO, et al. PKC regulates a farnesyl-electrostatic switch on K-Ras that promotes its association with Bcl-XL on mitochondria and induces apoptosis. Molecular Cell 2006;21:481–93.CrossRef
Fivaz, M, Meyer, T. Reversible intracellular translocation of KRas but not HRas in hippocampal neurons regulated by Ca2+/calmodulin. Journal of Cell Biology 2005;170:429–41.CrossRefGoogle Scholar
Sidhu, RS, Clough, RR, Bhullar, RP. Ca2+/calmodulin binds and dissociates K-RasB from membrane. Biochemical and Biophysical Research Communications 2003;304:655–60.CrossRef
Jura, N, Scotto-Lavino, E, Sobczyk, A, Bar-Sagi, D. Differential modification of Ras proteins by ubiquitination. Molecular Cell 2006;21:679–87.CrossRef
Rotblat, B, Ehrlich, M, Haklai, R, Kloog, Y. The Ras inhibitor farnesylthiosalicylic acid (Salirasib) disrupts the spatiotemporal localization of active Ras: a potential treatment for cancer. Methods in Enzymology 2008;439:467–89.CrossRef
Laheru, D, Shah, P, Rajeshkumar, NV, et al. Integrated preclinical and clinical development of S-trans, trans-farnesylthiosalicylic acid (FTS, Salirasib) in pancreatic cancer. Investigational New Drugs 2012;30:2391–9.CrossRef
Kamata, T, Feramisco, JR. Epidermal growth factor stimulates guanine nucleotide binding activity and phosphorylation of ras oncogene proteins. Nature 1984;310:147–50.CrossRef
Egan SE, Weinberg RA. The pathway to signal achievement. Nature 1993;365:781–3.CrossRef
Schulze, WX, Deng, L, Mann, M. Phosphotyrosine interactome of the ERBB-receptor kinase family. Molecular Systems Biology 2005;1:2005 0008.
Giubellino, A, Burke, TR, Bottaro, DP. Grb2 signaling in cell motility and cancer. Expert Opinion on Therapeutic Targets 2008;12:1021–33.CrossRef
Rojas, JM, Oliva, JL, Santos, E. Mammalian son of sevenless Guanine nucleotide exchange factors: old concepts and new perspectives. Genes and Cancer 2011;2:298–305.CrossRef
Ebinu, JO, Bottorff, DA, Chan, EY, et al. RasGRP, a Ras guanyl nucleotide- releasing protein with calcium- and diacylglycerol-binding motifs. Science 1998;280:1082–6.CrossRef
Tognon, CE, Kirk, HE, Passmore, LA, et al. Regulation of RasGRP via a phorbol ester-responsive C1 domain. Molecular and Cellular Biology 1998;18:6995–7008.CrossRef
Harden, TK, Sondek, J. Regulation of phospholipase C isozymes by ras superfamily GTPases. Annual Review of Pharmacology and Toxicology 2006;46:355–79.CrossRef
Ballare, C, Uhrig, M, Bechtold, T, et al. Two domains of the progesterone receptor interact with the estrogen receptor and are required for progesterone activation of the c-Src/Erk pathway in mammalian cells. Molecular and Cellular Biology 2003;23:1994–2008.CrossRef
Migliaccio, A, Piccolo, D, Castoria, G, et al. Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO Journal 1998;17:2008–18.CrossRef
Hood, JD, Frausto, R, Kiosses, WB, Schwartz, MA, Cheresh, DA. Differential alphav integrin-mediated Ras-ERK signaling during two pathways of angiogenesis. Journal of Cell Biology 2003;162:933–43.CrossRefGoogle ScholarPubMed
Adachi, T, Pimentel, DR, Heibeck, T, et al. S-glutathiolation of Ras mediates redox-sensitive signaling by angiotensin II in vascular smooth muscle cells. Journal of Biological Chemistry 2004;279:29 857–62.CrossRefGoogle ScholarPubMed
Pylayeva-Gupta, Y, Grabocka, E, Bar-Sagi, D. RAS oncogenes: weaving a tumorigenic web. Nature Reviews Cancer 2011;11:761–74.CrossRef
Campbell, PM, Der, CJ. Oncogenic Ras and its role in tumor cell invasion and metastasis. Seminars in Cancer Biology 2004;14:105–14.CrossRef
Coleman, ML, Marshall, CJ, Olson, MF. RAS and RHO GTPases in G1-phase cell-cycle regulation. Nature Reviews Molecular and Cellular Biology 2004;5:355–66.CrossRef
Filmus, J, Robles, AI, Shi, W, et al. Induction of cyclin D1 overexpression by activated ras. Oncogene 1994;9:3627–33.
Diehl, JA, Cheng, M, Roussel, MF, Sherr, CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes and Development 1998;12:3499–511.CrossRef
Cox, AD, Der, CJ. The dark side of Ras: regulation of apoptosis. Oncogene 2003;22:8999–9006.CrossRef
Blancher, C, Moore, JW, Robertson, N, Harris, AL. Effects of ras and von Hippel-Lindau (VHL) gene mutations on hypoxia-inducible factor (HIF)-1alpha, HIF-2alpha, and vascular endothelial growth factor expression and their regulation by the phosphatidylinositol 3ʹ-kinase/Akt signaling pathway. Cancer Research 2001;61:7349–55.
Kranenburg, O, Gebbink, MF, Voest, EE. Stimulation of angiogenesis by Ras proteins. Biochimica et Biophysica Acta 2004;1654:23–37.CrossRef
Ancrile, BB, O’Hayer, KM, Counter, CM. Oncogenic ras-induced expression of cytokines: a new target of anti-cancer therapeutics. Molecular Interventions 2008;8:22–7.CrossRef
Gordon, M, Baksh, S. RASSF1A: not a prototypical Ras effector. Small GTPases 2011;2:148–57.CrossRef
Avruch, J, Xavier, R, Bardeesy, N, et al. Rassf family of tumor suppressor polypeptides. The Journal of Biological Chemistry 2009;284:11001–5.CrossRefGoogle ScholarPubMed
Donninger, H, Vos, MD, Clark, GJ. The RASSF1A tumor suppressor. Journal of Cell Science 2007;120:3163–72.CrossRefGoogle ScholarPubMed
DeNicola, GM, Tuveson, DA. RAS in cellular transformation and senescence. European Journal of Cancer 2009;45 Suppl 1:211–16.CrossRefGoogle ScholarPubMed
Benanti, JA, Galloway, DA. The normal response to RAS: senescence or transformation? Cell Cycle 2004;3:715–17.
Overmeyer, JH, Maltese, WA. Death pathways triggered by activated Ras in cancer cells. Frontiers in Bioscience 2011;16:1693–713.CrossRef
Matallanas, D, Romano, D, Al-Mulla, F, et al. Mutant K-Ras activation of the proapoptotic MST2 pathway is antagonized by wild-type K-Ras. Molecular Cell 2011;44:893–906.CrossRef
Chapman, PB, Hauschild, A, Robert, C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. New England Journal of Medicine 2011;364:2507–16.CrossRefGoogle ScholarPubMed
Bollag, G, Hirth, P, Tsai, J, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 2010;467:596–9.CrossRef
Guan, KL, Figueroa, C, Brtva, TR, et al. Negative regulation of the serine/threonine kinase B-Raf by Akt. Journal of Biological Chemistry 2000;275:27 354–9.Google ScholarPubMed
Zimmermann, S, Moelling, K. Phosphorylation and regulation of Raf by Akt (protein kinase B). Science 1999;286:1741–4.CrossRef
Pratilas, CA, Taylor, BS, Ye, Q, et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proceedings of the National Academy of Sciences USA 2009;106:4519–24.CrossRef
Nazarian, R, Shi, H, Wang, Q, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010;468:973–7.CrossRef
Johannessen, CM, Boehm, JS, Kim, SY, et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature 2010;468:968–72.CrossRef
Wagle, N, Emery, C, Berger, MF, et al. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. Journal of Clinical Oncology 2011;29:3085–96.CrossRefGoogle ScholarPubMed
Poulikakos, PI, Zhang, C, Bollag, G, Shokat, KM, Rosen, N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature 2010;464:427–30.CrossRef
Hatzivassiliou, G, Song, K, Yen, I, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 2010;464:431–5.CrossRef
Heidorn, SJ, Milagre, C, Whittaker, S, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 2010;140:209–21.CrossRef
Poulikakos, PI, Persaud, Y, Janakiraman, M, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature 2011;480:387–90.CrossRef
Riera, L, Lasorsa, E, Ambrogio, C, et al. Involvement of Grb2 adaptor protein in nucleophosmin-anaplastic lymphoma kinase (NPM-ALK)-mediated signaling and anaplastic large cell lymphoma growth. Journal of Biological Chemistry 2010;285:26 441–50.CrossRefGoogle ScholarPubMed
Chiarle, R, Voena, C, Ambrogio, C, Piva, R, Inghirami, G. The anaplastic lymphoma kinase in the pathogenesis of cancer. Nature Reviews Cancer 2008;8:11–23.CrossRef
Mendoza, MC, Er, EE, Blenis, J. The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends in Biochemical Sciences. 2011;36:320–8.CrossRef
Castellano, E, Downward, J. RAS interaction with PI3K: more than just another effector pathway. Genes and Cancer 2011;2:261–74.CrossRef
Kolch, W. Meaningful relationships: the regulation of the Ras/Raf/MEK/ERK pathway by protein interactions. Biochemical Journal 2000;351 Pt 2:289–305.
Ceresa, BP, Horvath, CM, Pessin, JE. Signal transducer and activator of transcription-3 serine phosphorylation by insulin is mediated by a Ras/Raf/MEK-dependent pathway. Endocrinology 1997;138:4131–7.CrossRef
Debidda, M, Wang, L, Zang, H, Poli, V, Zheng, Y. A role of STAT3 in Rho GTPase-regulated cell migration and proliferation. Journal of Biological Chemistry 2005;280:17 275–85.CrossRefGoogle ScholarPubMed
Simon, AR, Vikis, HG, Stewart, S, et al. Regulation of STAT3 by direct binding to the Rac1 GTPase. Science 2000;290:144–7.CrossRef
Wu, JC, Chen, TY, Yu, CT, et al. Identification of V23RalA-Ser194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. Journal of Biological Chemistry 2005;280:9013–22.CrossRefGoogle ScholarPubMed
Lim, KH, Brady, DC, Kashatus, DF, et al. Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA. Molecular and Cellular Biology 2010;30:508–23.CrossRef
Mechta, F, Lallemand, D, Pfarr, CM, Yaniv, M. Transformation by ras modifies AP1 composition and activity. Oncogene 1997;14:837–47.CrossRef
Norris, JL, Baldwin, AS. Oncogenic Ras enhances NF-kappaB transcriptional activity through Raf-dependent and Raf-independent mitogen-activated protein kinase signaling pathways. Journal of Biological Chemistry 1999;274:13 841–6.CrossRefGoogle ScholarPubMed
Finco, TS, Westwick, JK, Norris, JL, et al. Oncogenic Ha-Ras-induced signaling activates NF-kappaB transcriptional activity, which is required for cellular transformation. Journal of Biological Chemistry 1997;272:24 113–6.CrossRefGoogle ScholarPubMed
Yang, JY, Zong, CS, Xia, W, et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nature Cell Biology 2008;10:138–48.CrossRef
Sears, R, Leone, G, DeGregori, J, Nevins, JR. Ras enhances Myc protein stability. Molecular Cell 1999;3:169–79.CrossRef
Sears, R, Nuckolls, F, Haura, E, et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes and Development 2000;14:2501–14.CrossRef
Bachireddy, P, Bendapudi, PK, Felsher, DW. Getting at MYC through RAS. Clinical Cancer Research 2005;11:4278–81.CrossRef
Land, H, Parada, LF, Weinberg, RA. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 1983;304:596–602.CrossRef
Sinn, E, Muller, W, Pattengale, P, et al. Coexpression of MMTV/v-Ha-ras and MMTV/c-myc genes in transgenic mice: synergistic action of oncogenes in vivo. Cell 1987;49:465–75.CrossRef
Huber, MA, Kraut, N, Beug, H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Current Opinion in Cell Biology 2005;17:548–58.CrossRef
Jang, JW, Boxer, RB, Chodosh, LA. Isoform-specific ras activation and oncogene dependence during MYC- and Wnt-induced mammary tumorigenesis. Molecular and Cellular Biology 2006;26:8109–21.CrossRef
Morris, JPt, Wang, SC, Hebrok, M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nature Reviews Cancer 2010;10:683–95.CrossRef
Lauth M. RAS and Hedgehog–partners in crime. Frontiers in Bioscience 2012;17:2259–70.
Schnidar, H, Eberl, M, Klingler, S, et al. Epidermal growth factor receptor signaling synergizes with Hedgehog/GLI in oncogenic transformation via activation of the MEK/ERK/JUN pathway. Cancer Research 2009;69:1284–92.CrossRef
Stecca, B, Mas, C, Clement, V, et al. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proceedings of the National Academy of Sciences USA 2007;104:5895–900.CrossRef
Thayer, SP, di Magliano, MP, Heiser, PW, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003;425:851–6.CrossRef
Mazur, PK, Einwachter, H, Lee, M, et al. Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proceedings of the National Academy of Sciences USA 2010;107:13 438–43.
Reiner, DJ. Ras effector switching as a developmental strategy. Small GTPases 2011;2:109–12.CrossRef
Sundaram, MV. The love-hate relationship between Ras and Notch. Genes and Development 2005;19:1825–39.CrossRef
Johnson, L, Greenbaum, D, Cichowski, K, et al. K-ras is an essential gene in the mouse with partial functional overlap with N-ras. Genes and Development 1997;11:2468–81.CrossRef
Perez de, Castro I, Diaz, R, Malumbres, M, et al. Mice deficient for N-ras: impaired antiviral immune response and T-cell function. Cancer Research 2003;63:1615–22.
Sparmann, A, Bar-Sagi, D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 2004;6:447–58.CrossRef
Johnson, DS, Chen, YH. Ras family of small GTPases in immunity and inflammation. Current Opinion in Pharmacology 2012;12:458–63.CrossRef
Hingorani, SR, Petricoin, EF, Maitra, A, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003;4:437–50.CrossRef
Bardeesy, N, Aguirre, AJ, Chu, GC, et al. Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proceedings of the National Academy of Sciences USA 2006;103:5947–52.CrossRef
Bardeesy, N, Cheng, KH, Berger, JH, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes and Development 2006;20:3130–46.CrossRef
Hingorani, SR, Wang, L, Multani, AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005;7:469–83.CrossRef
Shaw, AT, Meissner, A, Dowdle, JA, et al. Sprouty-2 regulates oncogenic K-ras in lung development and tumorigenesis. Genes and Development 2007;21:694–707.CrossRef
Velho, S, Haigis, KM. Regulation of homeostasis and oncogenesis in the intestinal epithelium by Ras. Experimental Cell Research 2011;317:2732–9.CrossRef
Johnson, L, Mercer, K, Greenbaum, D, et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 2001;410:1111–16.CrossRef
Kissil, JL, Walmsley, MJ, Hanlon, L, et al. Requirement for Rac1 in a K-ras induced lung cancer in the mouse. Cancer Research 2007;67:8089–94.CrossRef
Cellurale, C, Sabio, G, Kennedy, NJ, et al. Requirement of c-Jun NH(2)-terminal kinase for Ras-initiated tumor formation. Molecular and Cellular Biology 2011;31:1565–76.CrossRef
Charlin, A, Vexliard, D, Guerre, D, Peuteuil, P. [Severe retardation of affective origin, curable by environmental psychotherapy: ethiological approach]. Annals of Medical Psychology (Paris) 1975;1:356–62.
Der, CJ, Finkel, T, Cooper, GM. Biological and biochemical properties of human rasH genes mutated at codon 61. Cell 1986;44:167–76.CrossRef
Ahrendt, SA, Decker, PA, Alawi, EA, et al. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer 2001;92:1525–30.3.0.CO;2-H>CrossRef
Riely, GJ, Kris`, MG, Rosenbaum, D, et al. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clinical Cancer Research 2008;14:5731–4.CrossRef
Porta, M, Crous-Bou, M, Wark, PA, et al. Cigarette smoking and K-ras mutations in pancreas, lung and colorectal adenocarcinomas: etiopathogenic similarities, differences and paradoxes. Mutation Research 2009;682:83–93.CrossRef
Al-Mulla, F, Milner-White, EJ, Going, JJ, Birnie, GD. Structural differences between valine-12 and aspartate-12 Ras proteins may modify carcinoma aggression. Journal of Pathology 1999;187:433–8.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
Forrester, K, Almoguera, C, Han, K, Grizzle, WE, Perucho, M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature. 1987;327:298–303.CrossRef
Capella, G, Cronauer-Mitra, S, Pienado, MA, Perucho, M. Frequency and spectrum of mutations at codons 12 and 13 of the c-K-ras gene in human tumors. Environmental Health Perspectives 1991;93:125–31.CrossRef
Edkins, S, O’Meara, S, Parker, A, et al. Recurrent KRAS codon 146 mutations in human colorectal cancer. Cancer Biology and Therapy 2006;5:928–32.CrossRef
Jones, S, Zhang, X, Parsons, DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008;321:1801–6.CrossRef
Chin, L, Tam, A, Pomerantz, J, et al. Essential role for oncogenic Ras in tumour maintenance. Nature 1999;400:468–72.CrossRef
Chapman, PB, Hauschild, A, Robert, C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. New England Journal of Medicine 2011;364:2507–16.CrossRefGoogle ScholarPubMed
Greger, JG, Eastman, SD, Zhang, V, et al. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Molecular Cancer Therapeutics 2012;11:909–20.CrossRef
Tidyman, WE, Rauen, KA. The RASopathies: developmental syndromes of Ras/MAPK pathway dysregulation. Current Opinion in Genetic Development 2009;19:230–6.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Ras
    • By Adrienne D. Cox, Departments of Radiation Oncology and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, Molly J. DeCristo, Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.022
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Ras
    • By Adrienne D. Cox, Departments of Radiation Oncology and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, Molly J. DeCristo, Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.022
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Ras
    • By Adrienne D. Cox, Departments of Radiation Oncology and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, Molly J. DeCristo, Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.022
Available formats
×