Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Ras superfamily GEFs and GAPs: validated and tractable targets for cancer therapy?

Key Points

  • There is increasing evidence that the aberrant activity of numerous members of the Ras superfamily of small GTPases contributes to cancer growth, invasion and metastasis.

  • Unlike the frequent direct mutational activation of the three Ras proteins (which occurs in 33% of human cancers), other Ras superfamily GTPases are deregulated by indirect mechanisms, commonly involving the altered expression or activity of their regulatory proteins.

  • Guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) that control the GDP–GTP cycling of specific members of the Ras superfamily have been shown to contribute to cancer by either promoting or suppressing tumour progression and growth.

  • GEFs and GAPs are deregulated in cancer by somatic mutation, changes in gene expression and through post-translational mechanisms owing to aberrant signalling caused by alterations in upstream oncogene or tumour suppressor function.

  • Although GEFs and GAPs are not considered classically druggable targets, there is growing evidence that supports the feasibility of targeting them. For example, nature has provided examples (such as brefeldin A) that provide proof-of-principle of GEF and GAP druggability.

  • The multi-domain structures of GEFs and GAPs contribute to their regulation by diverse signalling mechanisms and might also identify therapeutic approaches for pharmacological regulation of GEF and GAP activity in cancer.

Abstract

There is now considerable and increasing evidence for a causal role for aberrant activity of the Ras superfamily of small GTPases in human cancers. These GTPases function as GDP–GTP-regulated binary switches that control many fundamental cellular processes. A common mechanism of GTPase deregulation in cancer is the deregulated expression and/or activity of their regulatory proteins, guanine nucleotide exchange factors (GEFs) that promote formation of the active GTP-bound state and GTPase-activating proteins (GAPs) that return the GTPase to its GDP-bound inactive state. In this Review, we assess the association of GEFs and GAPs with cancer and their druggability for cancer therapeutics.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: GEFs and GAPs are multi-domain proteins.
Figure 2: Candidate anti-Ras inhibitors.
Figure 3: Regulators and effectors of Ras.
Figure 4: Regulation of RhoGEF activity.
Figure 5: Inhibition of GEFs by brefeldin A and related molecules.
Figure 6: Signalling networks regulated by Ras and Rho family GTPases in cancer.

Similar content being viewed by others

References

  1. Colicelli, J. Human RAS superfamily proteins and related GTPases. Sci. STKE 2004, RE13 (2004).

    PubMed  PubMed Central  Google Scholar 

  2. Wennerberg, K., Rossman, K. L. & Der, C. J. The Ras superfamily at a glance. J. Cell Sci. 118, 843–846 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Tcherkezian, J. & Lamarche-Vane, N. Current knowledge of the large RhoGAP family of proteins. Biol. Cell 99, 67–86 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Bos, J. L., Rehmann, H. & Wittinghofer, A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 129, 865–877 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Cox, A. D. & Der, C. J. Ras history: the saga continues. Small GTPases 1, 2–27 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Karnoub, A. E. & Weinberg, R. A. Ras oncogenes: split personalities. Nature Rev. Mol. Cell Biol. 9, 517–531 (2008).

    Article  CAS  Google Scholar 

  7. Ma, W. W. & Adjei, A. A. Novel agents on the horizon for cancer therapy. CA Cancer J. Clin. 59, 111–137 (2009).

    Article  PubMed  Google Scholar 

  8. Feuerstein, J., Kalbitzer, H. R., John, J., Goody, R. S. & Wittinghofer, A. Characterisation of the metal-ion-GDP complex at the active sites of transforming and nontransforming p21 proteins by observation of the 17O-Mn superhyperfine coupling and by kinetic methods. Eur. J. Biochem. 162, 49–55 (1987).

    Article  CAS  PubMed  Google Scholar 

  9. Blum, R., Cox, A. D. & Kloog, Y. Inhibitors of chronically active ras: potential for treatment of human malignancies. Recent Pat. Anticancer Drug Discov. 3, 31–47 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Roberts, P. J. & Der, C. J. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 26, 3291–3310 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Ha, V. L., Luo, R., Nie, Z. & Randazzo, P. A. Contribution of AZAP-Type Arf GAPs to cancer cell migration and invasion. Adv. Cancer Res. 101, 1–28 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. D'Souza-Schorey, C. & Chavrier, P. ARF proteins: roles in membrane traffic and beyond. Nature Rev. Mol. Cell Biol. 7, 347–358 (2006).

    Article  CAS  Google Scholar 

  13. Sjoblom, T. et al. The consensus coding sequences of human breast and colorectal cancers. Science 314, 268–274 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Wood, L. D. et al. The genomic landscapes of human breast and colorectal cancers. Science 318, 1108–1113 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Srivastava, S. K., Wheelock, R. H., Aaronson, S. A. & Eva, A. Identification of the protein encoded by the human diffuse B-cell lymphoma (dbl) oncogene. Proc. Natl Acad. Sci. USA 83, 8868–8872 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Katzav, S., Martin-Zanca, D. & Barbacid, M. vav, a novel human oncogene derived from a locus ubiquitously expressed in hematopoietic cells. EMBO J. 8, 2283–2290 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Miki, T., Smith, C. L., Long, J. E., Eva, A. & Fleming, T. P. Oncogene ECT2 is related to regulators of small GTP-binding proteins. Nature 362, 462–465 (1993).

    Article  CAS  PubMed  Google Scholar 

  20. Whitehead, I., Kirk, H. & Kay, R. Expression cloning of oncogenes by retroviral transfer of cDNA libraries. Mol. Cell. Biol. 15, 704–710 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ebinu, J. O. et al. RasGRP, a Ras guanyl nucleotide- releasing protein with calcium- and diacylglycerol-binding motifs. Science 280, 1082–1086 (1998). This study provided an important demonstration of an alternative mode of RasGEF regulation, by diacylglycerol-mediated recruitment to the plasma membrane. As PLC isoforms function downstream of cell surface receptors that are aberrantly activated in cancer, this provided another example of how Ras may be activated through the actions of a GEF.

    Article  CAS  PubMed  Google Scholar 

  22. Tognon, C. E. et al. Regulation of RasGRP via a phorbol ester-responsive C1 domain. Mol. Cell. Biol. 18, 6995–7008 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Reuther, G. W. et al. RasGRP4 is a novel Ras activator isolated from acute myeloid leukemia. J. Biol. Chem. 277, 30508–30514 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Whitehead, I. P., Campbell, S., Rossman, K. L. & Der, C. J. Dbl family proteins. Biochim. Biophys. Acta 1332, F1–F23 (1997).

  25. Prenen, H., Tejpar, S. & Cutsem, E. V. New strategies for treatment of KRAS mutant metastatic colorectal cancer. Clin. Cancer Res. 16, 2921–2926 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Blumberg, P. M. et al. Wealth of opportunity - the C1 domain as a target for drug development. Curr. Drug Targets 9, 641–652 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Margarit, S. M. et al. Structural evidence for feedback activation by Ras.GTP of the Ras-specific nucleotide exchange factor SOS. Cell 112, 685–695 (2003). This study provided structural evidence for a feed-forward mechanism in which activated Ras can then activate a RasGEF.

    Article  CAS  PubMed  Google Scholar 

  28. Roberts, A. E. et al. Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nature Genet. 39, 70–74 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Tartaglia, M. et al. Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nature Genet. 39, 75–79 (2007). References 28 and 29 identified mutationally activated SOS1 in a developmental disorder in which other components of Ras signalling are also mutationally activated.

    Article  CAS  PubMed  Google Scholar 

  30. Swanson, K. D. et al. SOS1 mutations are rare in human malignancies: implications for Noonan Syndrome patients. Genes Chromosomes Cancer 47, 253–259 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Tidyman, W. E. & Rauen, K. A. The RASopathies: developmental syndromes of Ras/MAPK pathway dysregulation. Curr. Opin. Genet. Dev. 19, 230–236 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lopez, I., Mak, E. C., Ding, J., Hamm, H. E. & Lomasney, J. W. A novel bifunctional phospholipase c that is regulated by Gα 12 and stimulates the Ras/mitogen-activated protein kinase pathway. J. Biol. Chem. 276, 2758–2765 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Song, C. et al. Regulation of a novel human phospholipase C, PLCɛ, through membrane targeting by Ras. J. Biol. Chem. 276, 2752–2757 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Kelley, G. G., Reks, S. E., Ondrako, J. M. & Smrcka, A. V. Phospholipase Cɛ: a novel Ras effector. EMBO J. 20, 743–754 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ikuta, S., Edamatsu, H., Li, M., Hu, L. & Kataoka, T. Crucial role of phospholipase C ɛ in skin inflammation induced by tumor-promoting phorbol ester. Cancer Res. 68, 64–72 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Song, C. et al. Differential roles of Ras and Rap1 in growth factor-dependent activation of phospholipase Cɛ. Oncogene 21, 8105–8113 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Bodemann, B. O. & White, M. A. Ral GTPases and cancer: linchpin support of the tumorigenic platform. Nature Rev. Cancer 8, 133–140 (2008).

    Article  CAS  Google Scholar 

  38. Gonzalez-Garcia, A. et al. RALGDS is required for tumor formation in a model of skin carcinogenesis. Cancer Cell 7, 219–226 (2005). This paper demonstrated a crucial role for a RalGEF in a mouse model of Ras-induced skin cancer.

    Article  CAS  PubMed  Google Scholar 

  39. Vigil, D. et al. Aberrant overexpression of the RGL2 ral small GTPase-specific guanine nucleotide exchange factor promotes pancreatic cancer growth through ral-dependent and -independent mechanisms. J. Biol. Chem. 27 Aug 2010 (doi:10.1074/jbc.M110.116756).

  40. Chien, Y. & White, M. A. RAL GTPases are linchpin modulators of human tumour-cell proliferation and survival. EMBO Rep. 4, 800–806 (2003). This study showed that the two highly related substrates of RalGEFs may have highly divergent roles in normal and neoplastic cell growth and survival.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yin, J. et al. Activation of the RalGEF/Ral pathway promotes prostate cancer metastasis to bone. Mol. Cell. Biol. 27, 7538–7550 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Oxford, G. et al. RALA and RALB: antagonistic relatives in cancer cell migration. Cancer Res. 65, 7111–7120 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Lim, K. H. et al. Divergent roles for RALA and RALB in malignant growth of human pancreatic carcinoma cells. Curr. Biol. 16, 2385–2394 (2006). This study showed that a RalGEF effector pathway may be a crucial mechanism for Ras-mediated oncogenesis, in particular in pancreatic cancer.

    Article  CAS  PubMed  Google Scholar 

  44. Vega, F. M. & Ridley, A. J. Rho GTPases in cancer cell biology. FEBS Lett. 582, 2093–2101 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Ellenbroek, S. I. & Collard, J. G. Rho GTPases: functions and association with cancer. Clin. Exp. Metastasis 24, 657–672 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Fernandez-Zapico, M. E. et al. Ectopic expression of VAV1 reveals an unexpected role in pancreatic cancer tumorigenesis. Cancer Cell 7, 39–49 (2005). This study found a crucial role for a RacGEF that is overexpressed in pancreatic cancer and activated by EGFR.

    Article  CAS  PubMed  Google Scholar 

  47. Patel, V. et al. Persistent activation of RAC1 in squamous carcinomas of the head and neck: evidence for an EGFR/VAV2 signaling axis involved in cell invasion. Carcinogenesis 28, 1145–1152 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Salhia, B. et al. The guanine nucleotide exchange factors TRIO, ECT2, and VAV3 mediate the invasive behavior of glioblastoma. Am. J. Pathol. 173, 1828–1838 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Brantley-Sieders, D. M. et al. Host deficiency in VAV2/3 guanine nucleotide exchange factors impairs tumor growth, survival, and angiogenesis in vivo. Mol. Cancer Res. 7, 615–623 (2009). The study found that a deficiency of VAV2 and VAV3 in normal mouse tissue impaired the growth of transplanted cancer cells, demonstrating a crucial role for some RhoGEFs in the tumour microenvironment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Qin, J. et al. Upregulation of PIP3-dependent Rac exchanger 1 (P-Rex1) promotes prostate cancer metastasis. Oncogene 28, 1853–1863 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Karlsson, R., Pedersen, E. D., Wang, Z. & Brakebusch, C. Rho GTPase function in tumorigenesis. Biochim. Biophys. Acta 1796, 91–98 (2009).

    CAS  PubMed  Google Scholar 

  52. Shields, J. M. et al. Lack of extracellular signal-regulated kinase mitogen-activated protein kinase signaling shows a new type of melanoma. Cancer Res. 67, 1502–1512 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Fine, B. et al. Activation of the PI3K pathway in cancer through inhibition of PTEN by exchange factor P-REX2a. Science 325, 1261–1265 (2009). This paper identified a function for a RacGEF that is distinct from its activation of Rac, in which direct association with the PTEN tumour suppressor caused activation of PI3K signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tatsumoto, T., Xie, X., Blumenthal, R., Okamoto, I. & Miki, T. Human ECT2 is an exchange factor for Rho GTPases, phosphorylated in G2/M phases, and involved in cytokinesis. J. Cell Biol. 147, 921–928 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Solski, P. A. et al. Requirement for C-terminal sequences in regulation of ECT2 guanine nucleotide exchange specificity and transformation. J. Biol. Chem. 279, 25226–25233 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Saito, S. et al. Rho exchange factor ECT2 is induced by growth factors and regulates cytokinesis through the N-terminal cell cycle regulator-related domains. J. Cell. Biochem. 90, 819–836 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Sano, M. et al. Expression level of ECT2 proto-oncogene correlates with prognosis in glioma patients. Oncol. Rep. 16, 1093–1098 (2006).

    CAS  PubMed  Google Scholar 

  58. Hirata, D. et al. Involvement of epithelial cell transforming sequence-2 oncoantigen in lung and esophageal cancer progression. Clin. Cancer Res. 15, 256–266 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Zhang, M. L., Lu, S., Zhou, L. & Zheng, S. S. Correlation between ECT2 gene expression and methylation change of ECT2 promoter region in pancreatic cancer. Hepatobiliary Pancreat Dis. Int. 7, 533–538 (2008).

    CAS  PubMed  Google Scholar 

  60. Justilien, V. & Fields, A. P. ECT2 links the PKCiota-Par6α complex to RAC1 activation and cellular transformation. Oncogene 28, 3597–3607 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kourlas, P. J. et al. Identification of a gene at 11q23 encoding a guanine nucleotide exchange factor: evidence for its fusion with MLL in acute myeloid leukemia. Proc. Natl Acad. Sci. USA 97, 2145–2150 (2000). This paper identified a rearranged gene in an acute myelogenous leukaemia that encoded a chimeric protein that altered the structure of a RhoGEF.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kelly, P. et al. The G12 family of heterotrimeric G proteins promotes breast cancer invasion and metastasis. Proc. Natl Acad. Sci. USA 103, 8173–8178 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kelly, P. et al. A role for the G12 family of heterotrimeric G proteins in prostate cancer invasion. J. Biol. Chem. 281, 26483–26490 (2006).

    Article  CAS  PubMed  Google Scholar 

  64. Sahay, S. et al. The RhoGEF domain of p210 Bcr-Abl activates RHOA and is required for transformation. Oncogene 27, 2064–2071 (2008). This paper found that the BCR–ABL1 oncoprotein may promote oncogenesis, partly through its RhoGEF domain and activation of Rho GTPase.

    Article  CAS  PubMed  Google Scholar 

  65. Yoshizuka, N. et al. An alternative transcript derived from the TRIO locus encodes a guanosine nucleotide exchange factor with mouse cell-transforming potential. J. Biol. Chem. 279, 43998–44004 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Lambert, J. M. et al. TIAM1 mediates Ras activation of Rac by a PI3K-independent mechanism. Nature Cell Biol. 4, 621–625 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Malliri, A. et al. Mice deficient in the Rac activator TIAM1 are resistant to Ras-induced skin tumours. Nature 417, 867–871 (2002). This study found that the loss of the TIAM1 RacGEF impeded mutant Ras-induced skin tumour formation, but promoted later-stage malignant conversion, demonstrating that GEFs may possess stage-specific oncogenic or tumour suppressive functions.

    Article  CAS  PubMed  Google Scholar 

  68. Malliri, A. et al. The rac activator TIAM1 is a Wnt-responsive gene that modifies intestinal tumor development. J. Biol. Chem. 281, 543–548 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Strumane, K., Rygiel, T., van der Valk, M. & Collard, J. G. TIAM1-deficiency impairs mammary tumor formation in MMTV-c-neu but not in MMTV-c-myc mice. J. Cancer Res. Clin. Oncol. 135, 69–80 (2009).

    Article  CAS  PubMed  Google Scholar 

  70. Stebel, A., Brachetti, C., Kunkel, M., Schmidt, M. & Fritz, G. Progression of breast tumors is accompanied by a decrease in expression of the Rho guanine exchange factor TIAM1. Oncol. Rep. 21, 217–222 (2009).

    CAS  PubMed  Google Scholar 

  71. Cote, J. F. & Vuori, K. Identification of an evolutionarily conserved superfamily of DOCK180-related proteins with guanine nucleotide exchange activity. J. Cell Sci. 115, 4901–4913 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Meller, N., Merlot, S. & Guda, C. CZH proteins: a new family of Rho-GEFs. J. Cell Sci. 118, 4937–4946 (2005).

    Article  CAS  PubMed  Google Scholar 

  73. Yang, J., Zhang, Z., Roe, S. M., Marshall, C. J. & Barford, D. Activation of Rho GTPases by DOCK exchange factors is mediated by a nucleotide sensor. Science 325, 1398–1402 (2009).

    Article  CAS  PubMed  Google Scholar 

  74. Jarzynka, M. J. et al. ELMO1 and Dock180, a bipartite RAC1 guanine nucleotide exchange factor, promote human glioma cell invasion. Cancer Res. 67, 7203–7211 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Gadea, G., Sanz-Moreno, V., Self, A., Godi, A. & Marshall, C. J. DOCK10-mediated CDC42 activation is necessary for amoeboid invasion of melanoma cells. Curr. Biol. 18, 1456–1465 (2008).

    Article  CAS  PubMed  Google Scholar 

  76. Sanz-Moreno, V. et al. Rac activation and inactivation control plasticity of tumor cell movement. Cell 135, 510–523 (2008). This study showed that mesenchymal tumour cell motility is promoted by DOCK10-mediated activation of Rac, whereas amoeboid tumour cell movement involves activation of the RacGAP ARHGAP22 to inactivate Rac.

    Article  CAS  PubMed  Google Scholar 

  77. Boulay, P. L., Cotton, M., Melancon, P. & Claing, A. ADP-ribosylation factor 1 controls the activation of the phosphatidylinositol 3-kinase pathway to regulate epidermal growth factor-dependent growth and migration of breast cancer cells. J. Biol. Chem. 283, 36425–36434 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Muralidharan-Chari, V. et al. ADP-ribosylation factor 6 regulates tumorigenic and invasive properties in vivo. Cancer Res. 69, 2201–2209 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Ahn, J. Y., Hu, Y., Kroll, T. G., Allard, P. & Ye, K. PIKE-A is amplified in human cancers and prevents apoptosis by up-regulating Akt. Proc. Natl Acad. Sci. USA 101, 6993–6998 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sabe, H. et al. The EGFR-GEP100-ARF6-AMAP1 signaling pathway specific to breast cancer invasion and metastasis. Traffic 10, 982–993 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Morishige, M. et al. GEP100 links epidermal growth factor receptor signalling to ARF6 activation to induce breast cancer invasion. Nature Cell Biol. 10, 85–92 (2008). This study found a clear role for a specific ArfGEF in breast cancer invasion and metastasis, and this ArfGEF directly links EGFR activation to Arf signalling.

    Article  CAS  PubMed  Google Scholar 

  82. Hashimoto, S. et al. Requirement for ARF6 in breast cancer invasive activities. Proc. Natl Acad. Sci. USA 101, 6647–6652 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, M. et al. EFA6A enhances glioma cell invasion through ADP ribosylation factor 6/extracellular signal-regulated kinase signaling. Cancer Res. 66, 1583–1590 (2006).

    Article  CAS  PubMed  Google Scholar 

  84. Yano, H. et al. Fbx8 makes ARF6 refractory to function via ubiquitination. Mol. Biol. Cell 19, 822–832 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. McClatchey, A. I. Neurofibromatosis. Annu. Rev. Pathol. 2, 191–216 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Brems, H., Beert, E., de Ravel, T. & Legius, E. Mechanisms in the pathogenesis of malignant tumours in neurofibromatosis type 1. Lancet Oncol. 10, 508–515 (2009).

    Article  CAS  PubMed  Google Scholar 

  87. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  88. McGillicuddy, L. T. et al. Proteasomal and genetic inactivation of the NF1 tumor suppressor in gliomagenesis. Cancer Cell 16, 44–54 (2009). This paper described proteasomal degradation of the RasGAP neurofibromin leading to Ras hyperactivation in cancer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Aspuria, P. J. & Tamanoi, F. The RHEB family of GTP-binding proteins. Cell. Signal 16, 1105–1112 (2004).

    Article  CAS  PubMed  Google Scholar 

  90. Napolioni, V. & Curatolo, P. Genetics and molecular biology of tuberous sclerosis complex. Curr. Genomics 9, 475–487 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Inoki, K. & Guan, K. L. Tuberous sclerosis complex, implication from a rare genetic disease to common cancer treatment. Hum. Mol. Genet. 18, R94–R100 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Yeung, R. S. et al. Predisposition to renal carcinoma in the Eker rat is determined by germ-line mutation of the tuberous sclerosis 2 (TSC2) gene. Proc. Natl Acad. Sci. USA 91, 11413–11416 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kobayashi, T., Hirayama, Y., Kobayashi, E., Kubo, Y. & Hino, O. A germline insertion in the tuberous sclerosis (Tsc2) gene gives rise to the Eker rat model of dominantly inherited cancer. Nature Genet. 9, 70–74 (1995).

    Article  PubMed  Google Scholar 

  94. Kwiatkowski, D. J. et al. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum. Mol. Genet. 11, 525–534 (2002).

    Article  CAS  PubMed  Google Scholar 

  95. Zhou, X. et al. DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RHOA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis. Int. J. Oncol. 32, 1285–1291 (2008).

    CAS  PubMed  Google Scholar 

  96. Lahoz, A. & Hall, A. DLC1: a significant GAP in the cancer genome. Genes Dev. 22, 1724–1730 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Yuan, B. Z. et al. Cloning, characterization, and chromosomal localization of a gene frequently deleted in human liver cancer (DLC-1) homologous to rat RhoGAP. Cancer Res. 58, 2196–2199 (1998).

    CAS  PubMed  Google Scholar 

  98. Xue, W. et al. DLC1 is a chromosome 8p tumor suppressor whose loss promotes hepatocellular carcinoma. Genes Dev. 22, 1439–1444 (2008). This study found that the rate of genetic loss of the RhoGAP DLC1 approaches that of the TP53 tumour suppressor in some cancers, and it demonstrated a clear role for RhoGAP loss in tumorigenesis in a mouse model of liver cancer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ching, Y. P. et al. Deleted in liver cancer (DLC) 2 encodes a RhoGAP protein with growth suppressor function and is underexpressed in hepatocellular carcinoma. J. Biol. Chem. 278, 10824–10830 (2003).

    Article  CAS  PubMed  Google Scholar 

  100. Nagaraja, G. M. & Kandpal, R. P. Chromosome 13q12 encoded Rho GTPase activating protein suppresses growth of breast carcinoma cells, and yeast two-hybrid screen shows its interaction with several proteins. Biochem. Biophys. Res. Commun. 313, 654–665 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. Durkin, M. E., Ullmannova, V., Guan, M. & Popescu, N. C. Deleted in liver cancer 3 (DLC-3), a novel Rho GTPase-activating protein, is downregulated in cancer and inhibits tumor cell growth. Oncogene 26, 4580–4589 (2007).

    Article  CAS  PubMed  Google Scholar 

  102. Scholz, R. P. et al. DLC1 interacts with 14-3-3 proteins to inhibit RhoGAP activity and block nucleocytoplasmic shuttling. J. Cell Sci. 122, 92–102 (2009).

    Article  CAS  PubMed  Google Scholar 

  103. Yang, X. Y. et al. p120Ras-GAP binds the DLC1 Rho-GAP tumor suppressor protein and inhibits its RHOA GTPase and growth-suppressing activities. Oncogene 28, 1401–1409 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Zhou, X., Thorgeirsson, S. S. & Popescu, N. C. Restoration of DLC-1 gene expression induces apoptosis and inhibits both cell growth and tumorigenicity in human hepatocellular carcinoma cells. Oncogene 23, 1308–1313 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Durkin, M. E. et al. DLC-1:a Rho GTPase-activating protein and tumour suppressor. J. Cell. Mol. Med. 11, 1185–1207 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Liao, Y. C. & Lo, S. H. Deleted in liver cancer-1 (DLC-1): a tumor suppressor not just for liver. Int. J. Biochem. Cell Biol. 40, 843–847 (2008).

    Article  CAS  PubMed  Google Scholar 

  107. Healy, K. D. et al. DLC-1 suppresses non-small cell lung cancer growth and invasion by RhoGAP-dependent and independent mechanisms. Mol. Carcinog. 47, 326–337 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Goodison, S. et al. The RhoGAP protein DLC-1 functions as a metastasis suppressor in breast cancer cells. Cancer Res. 65, 6042–6053 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Sabe, H., Onodera, Y., Mazaki, Y. & Hashimoto, S. ArfGAP family proteins in cell adhesion, migration and tumor invasion. Curr. Opin. Cell Biol. 18, 558–564 (2006).

    Article  CAS  PubMed  Google Scholar 

  110. Ahn, J. Y. et al. PIKE (phosphatidylinositol 3-kinase enhancer)-A GTPase stimulates Akt activity and mediates cellular invasion. J. Biol. Chem. 279, 16441–16451 (2004).

    Article  CAS  PubMed  Google Scholar 

  111. Cai, Y. et al. GGAP2/PIKE-a directly activates both the Akt and nuclear factor-κB pathways and promotes prostate cancer progression. Cancer Res. 69, 819–827 (2009).

    CAS  Google Scholar 

  112. Liu, X., Hu, Y., Hao, C., Rempel, S. A. & Ye, K. PIKE-A is a proto-oncogene promoting cell growth, transformation and invasion. Oncogene 26, 4918–4927 (2007).

    Article  CAS  PubMed  Google Scholar 

  113. Soundararajan, M., Yang, X., Elkins, J. M., Sobott, F. & Doyle, D. A. The centaurin γ-1 GTPase-like domain functions as an NTPase. Biochem. J. 401, 679–688 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Ehlers, J. P., Worley, L., Onken, M. D. & Harbour, J. W. DDEF1 is located in an amplified region of chromosome 8q and is overexpressed in uveal melanoma. Clin. Cancer Res. 11, 3609–3613 (2005).

    Article  CAS  PubMed  Google Scholar 

  115. Lin, D. et al. ASAP1, a gene at 8q24, is associated with prostate cancer metastasis. Cancer Res. 68, 4352–4359 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Onodera, Y. et al. Expression of AMAP1, an ArfGAP, provides novel targets to inhibit breast cancer invasive activities. EMBO J. 24, 963–973 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hashimoto, S. et al. Targeting AMAP1 and cortactin binding bearing an atypical src homology 3/proline interface for prevention of breast cancer invasion and metastasis. Proc. Natl Acad. Sci. USA 103, 7036–7041 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Okabe, H. et al. Isolation of development and differentiation enhancing factor-like 1 (DDEFL1) as a drug target for hepatocellular carcinomas. Int. J. Oncol. 24, 43–48 (2004).

    CAS  PubMed  Google Scholar 

  119. Ha, V. L. et al. ASAP3 is a focal adhesion-associated Arf GAP that functions in cell migration and invasion. J. Biol. Chem. 283, 14915–14926 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Wells, J. A. & McClendon, C. L. Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature 450, 1001–1009 (2007).

    Article  CAS  PubMed  Google Scholar 

  121. Goldberg, J. Structural basis for activation of ARF GTPase: mechanisms of guanine nucleotide exchange and GTP-myristoyl switching. Cell 95, 237–248 (1998).

    Article  CAS  PubMed  Google Scholar 

  122. Boriack-Sjodin, P. A., Margarit, S. M., Bar-Sagi, D. & Kuriyan, J. The structural basis of the activation of Ras by Sos. Nature 394, 337–343 (1998).

    Article  CAS  PubMed  Google Scholar 

  123. Worthylake, D. K., Rossman, K. L. & Sondek, J. Crystal structure of RAC1 in complex with the guanine nucleotide exchange region of TIAM1. Nature 408, 682–688 (2000).

    Article  CAS  PubMed  Google Scholar 

  124. Singleton, V. L., Bohonos, N. & Ullstrup, A. J. Decumbin, a new compound from a species of Penicillium. Nature 181, 1072–1073 (1958).

    Article  CAS  PubMed  Google Scholar 

  125. Peyroche, A. et al. Brefeldin A acts to stabilize an abortive ARF-GDP-Sec7 domain protein complex: involvement of specific residues of the Sec7 domain. Mol. Cell 3, 275–285 (1999).

    Article  CAS  PubMed  Google Scholar 

  126. Robineau, S., Chabre, M. & Antonny, B. Binding site of brefeldin A at the interface between the small G protein ADP-ribosylation factor 1 (ARF1) and the nucleotide-exchange factor Sec7 domain. Proc. Natl Acad. Sci. USA 97, 9913–9918 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Mossessova, E., Corpina, R. A. & Goldberg, J. Crystal structure of ARF1Sec7 complexed with Brefeldin A and its implications for the guanine nucleotide exchange mechanism. Mol. Cell 12, 1403–1411 (2003).

    Article  CAS  PubMed  Google Scholar 

  128. Renault, L., Guibert, B. & Cherfils, J. Structural snapshots of the mechanism and inhibition of a guanine nucleotide exchange factor. Nature 426, 525–530 (2003). References 127 and 128 provide an important structural snapshot of how a GEF inhibitor inhibits nucleotide exchange and provides a basis for the discovery of novel GEF inhibitors.

    Article  CAS  PubMed  Google Scholar 

  129. Donaldson, J. G., Finazzi, D. & Klausner, R. D. Brefeldin A inhibits Golgi membrane-catalysed exchange of guanine nucleotide onto ARF protein. Nature 360, 350–352 (1992).

    Article  CAS  PubMed  Google Scholar 

  130. Helms, J. B. & Rothman, J. E. Inhibition by brefeldin A of a Golgi membrane enzyme that catalyses exchange of guanine nucleotide bound to ARF. Nature 360, 352–354 (1992).

    Article  CAS  PubMed  Google Scholar 

  131. Zeeh, J. C. et al. Dual specificity of the interfacial inhibitor brefeldin a for arf proteins and sec7 domains. J. Biol. Chem. 281, 11805–11814 (2006).

    Article  CAS  PubMed  Google Scholar 

  132. Pommier, Y. & Cherfils, J. Interfacial inhibition of macromolecular interactions: nature's paradigm for drug discovery. Trends Pharmacol. Sci. 26, 138–145 (2005).

    Article  CAS  PubMed  Google Scholar 

  133. Viaud, J. et al. Structure-based discovery of an inhibitor of Arf activation by Sec7 domains through targeting of protein-protein complexes. Proc. Natl Acad. Sci. USA 104, 10370–10375 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Mayer, G. et al. Controlling small guanine-nucleotide-exchange factor function through cytoplasmic RNA intramers. Proc. Natl Acad. Sci. USA 98, 4961–4965 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Hafner, M. et al. Inhibition of cytohesins by SecinH3 leads to hepatic insulin resistance. Nature 444, 941–944 (2006).

    Article  CAS  PubMed  Google Scholar 

  136. Saenz, J. B. et al. Golgicide A reveals essential roles for GBF1 in Golgi assembly and function. Nature Chem. Biol. 5, 157–165 (2009).

    Article  CAS  Google Scholar 

  137. Schmidt, S., Diriong, S., Mery, J., Fabbrizio, E. & Debant, A. Identification of the first Rho-GEF inhibitor, TRIPα, which targets the RHOA-specific GEF domain of TRIO. FEBS Lett. 523, 35–42 (2002).

    Article  CAS  PubMed  Google Scholar 

  138. Bouquier, N. et al. Aptamer-derived peptides as potent inhibitors of the oncogenic RhoGEF Tgat. Chem. Biol. 16, 391–400 (2009).

    Article  CAS  PubMed  Google Scholar 

  139. Blangy, A. et al. Identification of TRIO-GEFD1 chemical inhibitors using the yeast exchange assay. Biol. Cell 98, 511–522 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Bouquier, N. et al. A cell active chemical GEF inhibitor selectively targets the TRIO/RhoG/RAC1 signaling pathway. Chem. Biol. 16, 657–666 (2009).

    Article  CAS  PubMed  Google Scholar 

  141. Evelyn, C. R. et al. High-throughput screening for small-molecule inhibitors of LARG-stimulated RHOA nucleotide binding via a novel fluorescence polarization assay. J. Biomol. Screen. 14, 161–172 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Gao, Y., Dickerson, J. B., Guo, F., Zheng, J. & Zheng, Y. Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc. Natl Acad. Sci. USA 101, 7618–7623 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ferri, N., Corsini, A., Bottino, P., Clerici, F. & Contini, A. Virtual screening approach for the identification of new RAC1 inhibitors. J. Med. Chem. 52, 4087–4090 (2009).

    Article  CAS  PubMed  Google Scholar 

  144. Gibbs, J. B., Schaber, M. D., Allard, W. J., Sigal, I. S. & Scolnick, E. M. Purification of ras GTPase activating protein from bovine brain. Proc. Natl Acad. Sci. USA 85, 5026–5030 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Scheffzek, K. et al. The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science 277, 333–338 (1997).

    Article  CAS  PubMed  Google Scholar 

  146. Roman, D. L. et al. Identification of small-molecule inhibitors of RGS4 using a high-throughput flow cytometry protein interaction assay. Mol. Pharmacol. 71, 169–175 (2007).

    Article  CAS  PubMed  Google Scholar 

  147. Hao, Y., Wong, R. & Feig, L. A. RALGDS couples growth factor signaling to Akt activation. Mol. Cell. Biol. 28, 2851–2859 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Gupta, S. et al. Binding of ras to phosphoinositide 3-kinase p110α is required for ras-driven tumorigenesis in mice. Cell 129, 957–968 (2007).

    Article  CAS  PubMed  Google Scholar 

  149. Vetter, I. R. & Wittinghofer, A. The guanine nucleotide-binding switch in three dimensions. Science 294, 1299–1304 (2001).

    Article  CAS  PubMed  Google Scholar 

  150. Mitin, N., Rossman, K. L. & Der, C. J. Signaling interplay in Ras superfamily function. Curr. Biol. 15, R563–R574 (2005).

    Article  CAS  PubMed  Google Scholar 

  151. Buday, L. & Downward, J. Many faces of Ras activation. Biochim. Biophys. Acta 1786, 178–187 (2008).

    CAS  PubMed  Google Scholar 

  152. Rossman, K. L., Der, C. J. & Sondek, J. GEF means go: turning on RHO GTPases with guanine nucleotide-exchange factors. Nature Rev. Mol. Cell Biol. 6, 167–180 (2005).

    Article  CAS  Google Scholar 

  153. Kahn, R. A. et al. Consensus nomenclature for the human ArfGAP domain-containing proteins. J. Cell Biol. 182, 1039–1044 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Aghazadeh, B., Lowry, W. E., Huang, X. Y. & Rosen, M. K. Structural basis for relief of autoinhibition of the Dbl homology domain of proto-oncogene Vav by tyrosine phosphorylation. Cell 102, 625–633 (2000). This paper described the activation mechanism for a RhoGEF by tyrosine phosphorylation, suggesting that protein kinase inhibitors may be one approach to block the activation of some RhoGEFs.

    Article  CAS  PubMed  Google Scholar 

  155. Llorca, O., Arias-Palomo, E., Zugaza, J. L. & Bustelo, X. R. Global conformational rearrangements during the activation of the GDP/GTP exchange factor VAV3. EMBO J. 24, 1330–1340 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Yohe, M. E. et al. Auto-inhibition of the Dbl family protein Tim by an N-terminal helical motif. J. Biol. Chem. 282, 13813–13823 (2007).

    Article  CAS  PubMed  Google Scholar 

  157. Yohe, M. E., Rossman, K. & Sondek, J. Role of the C-terminal SH3 domain and N-terminal tyrosine phosphorylation in regulation of Tim and related Dbl-family proteins. Biochemistry 47, 6827–6839 (2008).

    Article  CAS  PubMed  Google Scholar 

  158. Hart, M. J. et al. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Gα13. Science 280, 2112–2114 (1998). This study identified a RhoGEF downstream of GPCR signalling through Gα 13 , and established another indirect mechanism for Rho GTPase activation in cancer.

    Article  CAS  PubMed  Google Scholar 

  159. Booden, M. A., Siderovski, D. P. & Der, C. J. Leukemia-associated Rho guanine nucleotide exchange factor promotes Gα q-coupled activation of RHOA. Mol. Cell. Biol. 22, 4053–4061 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Suzuki, N. et al. Activation of leukemia-associated RhoGEF by Gα13 with significant conformational rearrangements in the interface. J. Biol. Chem. 284, 5000–5009 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kawasaki, Y. et al. Asef, a link between the tumor suppressor APC and G-protein signaling. Science 289, 1194–1197 (2000).

    Article  CAS  PubMed  Google Scholar 

  162. Murayama, K. et al. Crystal structure of the rac activator, Asef, reveals its autoinhibitory mechanism. J. Biol. Chem. 282, 4238–4242 (2007).

    Article  CAS  PubMed  Google Scholar 

  163. Mitin, N. et al. Release of autoinhibition of ASEF by APC leads to CDC42 activation and tumor suppression. Nature Struct. Mol. Biol. 14, 814–823 (2007).

    Article  CAS  Google Scholar 

  164. Sahai, E., Ishizaki, T., Narumiya, S. & Treisman, R. Transformation mediated by RHOA requires activity of ROCK kinases. Curr. Biol. 9, 136–145 (1999).

    Article  CAS  PubMed  Google Scholar 

  165. Sahai, E. & Marshall, C. J. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nature Cell Biol. 5, 711–719 (2003).

    Article  CAS  PubMed  Google Scholar 

  166. Croft, D. R. et al. Conditional ROCK activation in vivo induces tumor cell dissemination and angiogenesis. Cancer Res. 64, 8994–9001 (2004).

    Article  CAS  PubMed  Google Scholar 

  167. Wilkinson, S., Paterson, H. F. & Marshall, C. J. CDC42-MRCK and Rho-ROCK signalling cooperate in myosin phosphorylation and cell invasion. Nature Cell Biol. 7, 255–261 (2005).

    Article  CAS  PubMed  Google Scholar 

  168. Qu, J. et al. Activated PAK4 regulates cell adhesion and anchorage-independent growth. Mol. Cell. Biol. 21, 3523–3533 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Callow, M. G. et al. Requirement for PAK4 in the anchorage-independent growth of human cancer cell lines. J. Biol. Chem. 277, 550–558 (2002).

    Article  CAS  PubMed  Google Scholar 

  170. Chow, B. J. et al. Prognostic value of 64-slice cardiac computed tomography severity of coronary artery disease, coronary atherosclerosis, and left ventricular ejection fraction. J. Am. Coll. Cardiol. 55, 1017–1028 (2009).

    Article  Google Scholar 

  171. Kimmelman, A. C. et al. Genomic alterations link Rho family of GTPases to the highly invasive phenotype of pancreas cancer. Proc. Natl Acad. Sci. USA 105, 19372–19377 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. van der Horst, E. H. et al. Metastatic properties and genomic amplification of the tyrosine kinase gene ACK1. Proc. Natl Acad. Sci. USA 102, 15901–15906 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Mahajan, N. P., Whang, Y. E., Mohler, J. L. & Earp, H. S. Activated tyrosine kinase Ack1 promotes prostate tumorigenesis: role of Ack1 in polyubiquitination of tumor suppressor Wwox. Cancer Res. 65, 10514–10523 (2005).

    Article  CAS  PubMed  Google Scholar 

  174. Olson, M. F. Applications for ROCK kinase inhibition. Curr. Opin. Cell Biol. 20, 242–248 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Bivona, T. G. et al. PKC regulates a farnesyl-electrostatic switch on K-Ras that promotes its association with Bcl-XL on mitochondria and induces apoptosis. Mol. Cell 21, 481–493 (2006).

    Article  CAS  PubMed  Google Scholar 

  176. Lim, K. H. et al. Aurora-A phosphorylates, activates, and relocalizes the small GTPase RALA. Mol. Cell Biol. 30, 508–523 (2010).

    Article  CAS  PubMed  Google Scholar 

  177. Narumiya, S., Tanji, M. & Ishizaki, T. Rho signaling, ROCK and mDia1, in transformation, metastasis and invasion. Cancer Metastasis Rev. 28, 65–76 (2009).

    Article  CAS  PubMed  Google Scholar 

  178. van Dam, T. J., Rehmann, H., Bos, J. L. & Snel, B. Phylogeny of the CDC25 homology domain reveals rapid differentiation of Ras pathways between early animals and fungi. Cell. Signal 21, 1579–1585 (2009).

    Article  CAS  PubMed  Google Scholar 

  179. Raaijmakers, J. H. & Bos, J. L. Specificity in Ras and Rap signaling. J. Biol. Chem. 284, 10995–10999 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors' research on GEFs and GAPs is supported in part by grants number CA042978, CA129610, CA127152, CA67771 and CA106991 from the US National Institutes of Health (C.J.D.), from the American Cancer Society (D.V. and K.L.R.), and from the Centre National de la Recherche Scientifique, France, (J.C.), and by grants from the Association pour la Recherche Contre le Cancer, France, (J.C.) and the Agence Nationale de la Recherche, France (J.C.). The authors thank L. DeGraffenreid for outstanding assistance in the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Channing J. Der.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Related links

Related links

DATABASES

National Cancer Institute Drug Dictionary

Imatinib

Vinblastine

FURTHER INFORMATION

Channing Der's homepage

ClinicalTrials.gov

COSMIC database

Protein Data Bank

SMART

The Cancer Genome Project

Glossary

Ras GTPases

Key regulators of extracellular signal-regulated cytoplasmic signalling networks that control cell growth, survival and differentiation.

Rho

This subfamily of Ras GTPases shares similar roles in signal transduction to Ras GTPases and is best-characterized for the regulation of actin cytoskeletal organization and cell shape, movement and polarity.

Rab

This subfamily of Ras GTPases regulate membrane trafficking and intracellular transport.

Arf

This subfamily of Ras GTPases regulate membrane trafficking and intracellular transport.

Ran

This Ras GTPase regulates nucleocytoplasmic transport of macromolecules and the organization of the spindle apparatus during mitosis.

Noonan syndrome

This syndrome is characterized by short stature, characteristic facies, learning problems and a predisposition to develop leukaemia and other cancers, including myeloproliferative disease and neuroblastoma.

CDC25 homology domain

RasGEF catalytic domain, named after the first protein it was identified in: CDC25 in Saccharomyces cerevisiae.

Matrigel

The trade name for a gelatinous protein mixture that is secreted by mouse tumour cells. It resembles the complex extracellular environment found in many tissues and is commonly used as a three-dimensional matrix substrate for cell culture-based in vitro migration and invasion assays.

CAAX motif

C-terminal tetrapeptide sequence comprised of a cysteine, followed by two aliphatic amino acids and a terminal X residue that dictates specificity for farnesyltransferase or geranylgeranyltransferase-I catalysed addition of a C15 farnesyl or C20 geranylgeranyl isoprenoid lipid.

PAK

A group of six structurally similar human serine/threonine kinases that can function as effectors of Rac (PAK1–3) or CDC42 (PAK1–6).

Organotypic

Resembling an organ in vivo, either morphologically or functionally, or both.

Dbl homology (DH) domain

The RhoGEF catalytic domain, named after the first protein it was identified in, the Dbl protein encoded by a transforming gene identified from an NIH 3T3 focus formation assay using genomic DNA from a human diffuse B cell lymphoma.

Pleckstrin homology (PH) domain

A sequence of approximately 100 amino acids that is present in many signalling molecules and that commonly binds to phospholipids and proteins.

Philadelphia chromosome

The chromosome abnormality that causes chronic myeloid leukaemia, formed by a translocation between chromosomes 9 and 22, causing formation of the chimeric BCR–ABL1 tyrosine kinase.

Alternative splicing

A mechanism by which different forms of mature mRNAs are generated from the same gene, leading to the production of more than one related protein or isoform.

Invadopodia

Actin-rich subcellular protrusions with associated proteases used by carcinoma cells to degrade the extracellular matrix to promote invasion.

Farnesyltransferase

One of three human prenyltransferase enzymes, catalyses addition of a 15-carbon farnesyl group to proteins terminating with a CAAX tetrapeptide motif at the carboxyl terminus of a subset of Ras and Rho family GTPases.

Neurofibromatosis type 1

Patients with this autosomal dominant familial syndrome are at increased risk of developing tumours of the peripheral and central nervous system, including neurofibromas, plexiform neurofibromas, malignant peripheral nerve sheath tumours and low-grade gliomas.

mTOR

A serine/threonine kinase that regulates cell growth, cell proliferation, cell motility, cell survival, protein synthesis and gene transcription, autophagy and angiogenesis.

StAR-related lipid transfer (START) domain

A 200 amino acid motif initially identified as a lipid-binding domain.

Sterile α-motif

SAM. A 70 amino acid domain involved in protein–protein interactions that is found in a wide variety of proteins involved in many biological processes.

Druggability

The likelihood of being able to modulate the activity of a target protein with a small-molecule drug.

Sec7 domain

ArfGEF catalytic domain, named after the first protein that it was identified in S. cerevisiae SEC7.

Aptamer

A double-stranded DNA, single-stranded RNA or peptide that binds to specific molecular targets, such as a protein or metabolite. Aptamers are usually selected from large libraries of synthesized molecules.

Fluorescence polarization

A technique specially applied to study molecular interactions. When fluorescent molecules in solution are excited with plane-polarized light, they will rotate and tumble, and the planes into which light is emitted can be very different from the plane used for initial excitation.

RGS domain

Functions in a similar manner to GTPase-activating proteins by accelerating the intrinsic GTP hydrolysis activity of heterotrimeric Gα subunits, causing inactivation of GPCR signalling.

C1 zinc finger domain

An 50 amino acid phospholipid binding domain. It typically binds membrane-bound phorbol esters or diacylglycerol to promote membrane localization.

ROCK

A serine/threonine kinase that is an effector of RHOA and RHOC and phosphorylates proteins that regulate actin stress fibre formation and focal adhesion assembly.

MRCK

Serine/threonine kinases that preferentially bind to activated CDC42 and phosphorylate proteins that regulate actin reorganization.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vigil, D., Cherfils, J., Rossman, K. et al. Ras superfamily GEFs and GAPs: validated and tractable targets for cancer therapy?. Nat Rev Cancer 10, 842–857 (2010). https://doi.org/10.1038/nrc2960

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2960

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer