Skip to main content

Rho GTPases and Their Activators, Guanine Nucleotide Exchange Factors (GEFs): Their Roles in Glioma Cell Invasion

  • Chapter
  • First Online:
Signaling Pathways and Molecular Mediators in Metastasis

Abstract

Accumulated studies showed that constitutively activated Rho GTPases such as Rac1 in malignant human glioblastomas are responsible for the highly invasive phenotype observed in these aggressive brain cancers. Notably, no activating mutations of Rac1 have been reported in human glioblastomas or other types of cancers. Moreover, Rac1 and several GEFs have been implicated in cell invasion and metastasis of glioblastomas and other types of human cancers. Mechanistically, the functions of the Rho GTPases are regulated by three distinct classes of molecules. Among them, guanine nucleotide exchange factors (GEFs) are the activators for Rho GTPases. Here, we review the role of Rho GTPases, particularly Rac1 and several GEFs including Trio, SWAP-70, Ect2, Vav3 and Dock180-ELMO1 in ­glioblastoma cell invasion. Since studies of Rho GTPases and their GEFs in ­glioblastomas are just emerging, we place specific emphasis on the current knowledge of their roles in cell motility and cancer cell invasion as well as potential response to extracellular stimuli that promotes glioma cell invasion. Additionally, we include studies of two membrane receptor proteins, Fn14 and TROY that promote glioma cell invasion through activation of Rho GTPases. Lastly, we discuss future directions for understanding the functions of Rac1 and GEFs in glioma cell invasion and implications to establish these key modulators as potential targets to inhibit diffusely invasive glioblastomas in the brain. The insight provided by this review will help to develop new therapeutic approaches to treat patients with ­malignant gliomas.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

BAI-1:

Brain-specific angiogenesis inhibitor-1

Crk:

v-crk sarcoma virus CT10 oncogene homolog

Dbl:

Diffuse B-cell lypmphoma

DD:

Death domain

DH domain:

Dbl homology (DH) domain

DHR domain:

Dock-homology region

Dock:

Dedicator of cytokinesis

Ect2:

Epithelial cell transforming sequence 2

EGFR:

Epithelial growth factor receptor

ELMO1:

Engulfment and cell motility-1

Fn14:

Fibroblast growth factor inducible 14

GAP:

GTPase-activating protein

GBM:

Glioblastoma multiforme

GDI:

Guanine nucleotide dissociation inhibitor

GEF:

Guanine nucleotide exchange factor (GDP–GTP exchange factor)

p130Cas :

crk-associate substrate 130 kDa protein

PDGFR:

Platelet-derived growth factor receptor

PH domain:

Pleckstrin homology domain

PtdIns:

Phosphatidylinositol

Pyk:

proline-rich tyrosine kinase

RNAi:

RNA interference

ROCK:

Rho kinase

SH2 or 3:

src homologous domain 2 or 3

SWAP:

switching B-cell complex 70 kDa subunit

TNF:

Tumor necrosis factor

TNFRSF:

Tumor necrosis factor receptor superfamily

TRAF:

Tumor necrosis factor receptor associated factor

Trio:

Triple function domains (PTPRF interacting)

TROY/Tnfrsf19:

Tumor necrosis factor receptor superfamily member 19

TSC:

Triple sex combs

TWEAK:

Tumor necrosis factor-like weak inducer of apoptosis

Vav3:

vav 3 guanine nucleotide exchange factor

VEGFR:

Vascular endothelial growth factor receptor

References

  1. Wen PY, Kesari S (2008) Malignant gliomas in adults. N Engl J Med 359:492–507

    PubMed  CAS  Google Scholar 

  2. Furnari FB, Fenton T, Bachoo RM et al (2007) Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21:2683–2710

    PubMed  CAS  Google Scholar 

  3. Giese A, Bjerkvig R, Berens ME, Westphal M (2003) Cost of migration: invasion of malignant gliomas and implications for treatment. J Clin Oncol 21:1624–1636

    PubMed  CAS  Google Scholar 

  4. Lefranc F, Brotchi J, Kiss R (2005) Possible future issues in the treatment of glioblastomas: special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis. J Clin Oncol 23:2411–2422

    PubMed  CAS  Google Scholar 

  5. Tcga C (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:1061–1068

    Google Scholar 

  6. Burridge K, Wennerberg K (2004) Rho and Rac take center stage. Cell 116:167–179

    PubMed  CAS  Google Scholar 

  7. Schmitz AA, Govek EE, Bottner B, Van Aelst L (2000) Rho GTPases: signaling, migration, and invasion. Exp Cell Res 261:1–12

    PubMed  CAS  Google Scholar 

  8. Rossman KL, Der CJ, Sondek J (2005) GEF means go: turning on RHO GTPases with guanine nucleotide-exchange factors. Nat Rev Mol Cell Biol 6:167–180

    PubMed  CAS  Google Scholar 

  9. Eva A, Vecchio G, Rao CD, Tronick SR, Aaronson SA (1988) The predicted DBL oncogene ­product defines a distinct class of transforming proteins. Proc Natl Acad Sci U S A 85:2061–2065

    PubMed  CAS  Google Scholar 

  10. Schmidt S, Diriong S, Mery J, Fabbrizio E, Debant A (2002) Identification of the first Rho-GEF inhibitor, TRIPalpha, which targets the RhoA-specific GEF domain of Trio. FEBS Lett 523:35–42

    PubMed  CAS  Google Scholar 

  11. Moon SY, Zheng Y (2003) Rho GTPase-activating proteins in cell regulation. Trends Cell Biol 13:13–22

    PubMed  CAS  Google Scholar 

  12. DerMardirossian C, Bokoch GM (2005) GDIs: central regulatory molecules in Rho GTPase activation. Trends Cell Biol 15:356–363

    PubMed  CAS  Google Scholar 

  13. Dovas A, Couchman JR (2005) RhoGDI: multiple functions in the regulation of Rho family GTPase activities. Biochem J 390:1–9

    PubMed  CAS  Google Scholar 

  14. Schmidt A, Hall A (2002) Guanine nucleotide exchange factors for Rho GTPases: turning on the switch. Genes Dev 16:1587–1609

    PubMed  CAS  Google Scholar 

  15. Bernards A, Settleman J (2005) GAPs in growth factor signalling. Growth Factors 23:143–149

    PubMed  CAS  Google Scholar 

  16. Gomez del Pulgar T, Benitah SA, Valeron PF, Espina C, Lacal JC (2005) Rho GTPase expression in tumourigenesis: evidence for a significant link. Bioessays 27:602–613

    PubMed  Google Scholar 

  17. Etienne-Manneville S, Hall A (2002) Rho GTPases in cell biology. Nature 420:629–635

    PubMed  CAS  Google Scholar 

  18. Chan AY, Coniglio SJ, Chuang YY et al (2005) Roles of the Rac1 and Rac3 GTPases in human tumor cell invasion. Oncogene 24:7821–7829

    PubMed  CAS  Google Scholar 

  19. Raftopoulou M, Hall A (2004) Cell migration: Rho GTPases lead the way. Dev Biol 265:23–32

    PubMed  CAS  Google Scholar 

  20. Sahai E, Marshall CJ (2002) RHO-GTPases and cancer. Nat Rev Cancer 2:133–142

    PubMed  Google Scholar 

  21. Hall A (2005) Rho GTPases and the control of cell behaviour. Biochem Soc Trans 33:891–895

    PubMed  CAS  Google Scholar 

  22. Fernandez-Zapico ME, Gonzalez-Paz NC, Weiss E et al (2005) Ectopic expression of VAV1 reveals an unexpected role in pancreatic cancer tumorigenesis. Cancer Cell 7:39–49

    PubMed  CAS  Google Scholar 

  23. Bartolome RA, Molina-Ortiz I, Samaniego R, Sanchez-Mateos P, Bustelo XR, Teixido J (2006) Activation of Vav/Rho GTPase signaling by CXCL12 controls membrane-type matrix metalloproteinase-dependent melanoma cell invasion. Cancer Res 66:248–258

    PubMed  CAS  Google Scholar 

  24. Leeuwen FN, Kain HE, Kammen RA, Michiels F, Kranenburg OW, Collard JG (1997) The guanine nucleotide exchange factor Tiam1 affects neuronal morphology; opposing roles for the small GTPases Rac and Rho. J Cell Biol 139:797–807

    PubMed  CAS  Google Scholar 

  25. Minard ME, Ellis LM, Gallick GE (2006) Tiam1 regulates cell adhesion, migration and ­apoptosis in colon tumor cells. Clin Exp Metastasis 23:301–313

    PubMed  CAS  Google Scholar 

  26. Salhia B, Rutten F, Nakada M et al (2005) Inhibition of Rho-kinase affects astrocytoma morphology, motility, and invasion through activation of Rac1. Cancer Res 65:8792–8800

    PubMed  CAS  Google Scholar 

  27. Salhia B, Tran NL, Chan A et al (2008) The guanine nucleotide exchange factors trio, Ect2, and Vav3 mediate the invasive behavior of glioblastoma. Am J Pathol 173:1828–1838

    PubMed  CAS  Google Scholar 

  28. Hall A (2009) The cytoskeleton and cancer. Cancer Metastasis Rev 28:5–14

    PubMed  Google Scholar 

  29. Sahai E, Marshall CJ (2003) Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol 5:711–719

    PubMed  CAS  Google Scholar 

  30. Wolf K, Mazo I, Leung H et al (2003) Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J Cell Biol 160:267–277

    PubMed  CAS  Google Scholar 

  31. Sanz-Moreno V, Gadea G, Ahn J et al (2008) Rac activation and inactivation control plasticity of tumor cell movement. Cell 135:510–523

    PubMed  CAS  Google Scholar 

  32. Friedl P, Wolf K (2003) Tumour-cell invasion and migration: diversity and escape ­mechanisms. Nat Rev Cancer 3:362–374

    PubMed  CAS  Google Scholar 

  33. Friedl P, Wolf K (2010) Plasticity of cell migration: a multiscale tuning model. J Cell Biol 188:11–19

    PubMed  CAS  Google Scholar 

  34. Sanz-Moreno V, Marshall CJ (2010) The plasticity of cytoskeletal dynamics underlying ­neoplastic cell migration. Curr Opin Cell Biol 22:690–696

    PubMed  CAS  Google Scholar 

  35. Karlsson R, Pedersen ED, Wang Z, Brakebusch C (2009) Rho GTPase function in tumorigenesis. Biochim Biophys Acta 1796:91–98

    PubMed  CAS  Google Scholar 

  36. Harding MA, Theodorescu D (2010) RhoGDI signaling provides targets for cancer therapy. Eur J Cancer 46:1252–1259

    PubMed  CAS  Google Scholar 

  37. Preudhomme C, Roumier C, Hildebrand MP et al (2000) Nonrandom 4p13 rearrangements of the RhoH/TTF gene, encoding a GTP-binding protein, in non-Hodgkin’s lymphoma and multiple myeloma. Oncogene 19:2023–2032

    PubMed  CAS  Google Scholar 

  38. Rihet S, Vielh P, Camonis J, Goud B, Chevillard S, de Gunzburg J (2001) Mutation status of genes encoding RhoA, Rac1, and Cdc42 GTPases in a panel of invasive human colorectal and breast tumors. J Cancer Res Clin Oncol 127:733–738

    PubMed  CAS  Google Scholar 

  39. Fritz G, Brachetti C, Bahlmann F, Schmidt M, Kaina B (2002) Rho GTPases in human breast tumours: expression and mutation analyses and correlation with clinical parameters. Br J Cancer 87:635–644

    PubMed  CAS  Google Scholar 

  40. Ridley AJ (2004) Rho proteins and cancer. Breast Cancer Res Treat 84:13–19

    PubMed  CAS  Google Scholar 

  41. Iwadate Y, Sakaida T, Hiwasa T et al (2004) Molecular classification and survival prediction in human gliomas based on proteome analysis. Cancer Res 64:2496–2501

    PubMed  CAS  Google Scholar 

  42. Chuang YY, Tran NL, Rusk N, Nakada M, Berens ME, Symons M (2004) Role of synaptojanin 2 in glioma cell migration and invasion. Cancer Res 64:8271–8275

    PubMed  CAS  Google Scholar 

  43. Tran NL, McDonough WS, Savitch BA et al (2006) Increased fibroblast growth factor-­inducible 14 expression levels promote glioma cell invasion via Rac1 and nuclear factor-{kappa}B and correlate with poor patient outcome. Cancer Res 66:9535–9542

    PubMed  CAS  Google Scholar 

  44. Nakada M, Drake KL, Nakada S, Niska JA, Berens ME (2006) Ephrin-B3 ligand promotes glioma invasion through activation of Rac1. Cancer Res 66:8492–8500

    PubMed  CAS  Google Scholar 

  45. Gimona M, Buccione R, Courtneidge SA, Linder S (2008) Assembly and biological role of podosomes and invadopodia. Curr Opin Cell Biol 20:235–241

    PubMed  CAS  Google Scholar 

  46. Beadle C, Assanah MC, Monzo P, Vallee R, Rosenfeld SS, Canoll P (2008) The role of myosin II in glioma invasion of the brain. Mol Biol Cell 19:3357–3368

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  50. Cote JF, Motoyama AB, Bush JA, Vuori K (2005) A novel and evolutionarily conserved PtdIns(3,4,5)P3-binding domain is necessary for DOCK180 signalling. Nat Cell Biol 7:797–807

    PubMed  CAS  Google Scholar 

  51. Cote JF, Vuori K (2007) GEF what? Dock180 and related proteins help Rac to polarize cells in new ways. Trends Cell Biol 17:383–393

    PubMed  CAS  Google Scholar 

  52. Brugnera E, Haney L, Grimsley C et al (2002) Unconventional Rac-GEF activity is mediated through the Dock180-ELMO complex. Nat Cell Biol 4:574–582

    PubMed  CAS  Google Scholar 

  53. Meller N, Irani-Tehrani M, Ratnikov BI, Paschal BM, Schwartz MA (2004) The novel Cdc42 guanine nucleotide exchange factor, zizimin1, dimerizes via the Cdc42-binding CZH2 domain. J Biol Chem 279:37470–37476

    PubMed  CAS  Google Scholar 

  54. Schiller MR (2006) Coupling receptor tyrosine kinases to Rho GTPases–GEFs what’s the link. Cell Signal 18:1834–1843

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  56. Seol HJ, Smith CA, Salhia B, Rutka JT (2009) The guanine nucleotide exchange factor ­SWAP-70 modulates the migration and invasiveness of human malignant glioma cells. Transl Oncol 2:300–309

    PubMed  Google Scholar 

  57. Debant A, Serra-Pages C, Seipel K et al (1996) The multidomain protein Trio binds the LAR transmembrane tyrosine phosphatase, contains a protein kinase domain, and has separate rac-specific and rho-specific guanine nucleotide exchange factor domains. Proc Natl Acad Sci U S A 93:5466–5471

    PubMed  CAS  Google Scholar 

  58. Bateman J, Van Vactor D (2001) The Trio family of guanine-nucleotide-exchange factors: regulators of axon guidance. J Cell Sci 114:1973–1980

    PubMed  CAS  Google Scholar 

  59. Blangy A, Vignal E, Schmidt S, Debant A, Gauthier-Rouviere C, Fort P (2000) TrioGEF1 controls Rac- and Cdc42-dependent cell structures through the direct activation of rhoG. J Cell Sci 113(Pt 4):729–739

    PubMed  CAS  Google Scholar 

  60. Katoh H, Negishi M (2003) RhoG activates Rac1 by direct interaction with the Dock180-binding protein Elmo. Nature 424:461–464

    PubMed  CAS  Google Scholar 

  61. Briancon-Marjollet A, Ghogha A, Nawabi H et al (2008) Trio mediates netrin-1-induced Rac1 activation in axon outgrowth and guidance. Mol Cell Biol 28:2314–2323

    PubMed  CAS  Google Scholar 

  62. Charrasse S, Comunale F, Fortier M, Portales-Casamar E, Debant A, Gauthier-Rouviere C (2007) M-cadherin activates Rac1 GTPase through the Rho-GEF trio during myoblast fusion. Mol Biol Cell 18:1734–1743

    PubMed  CAS  Google Scholar 

  63. Lin MZ, Greenberg ME (2000) Orchestral maneuvers in the axon: trio and the control of axon guidance. Cell 101:239–242

    PubMed  CAS  Google Scholar 

  64. Portales-Casamar E, Briancon-Marjollet A, Fromont S, Triboulet R, Debant A (2006) Identification of novel neuronal isoforms of the Rho-GEF Trio. Biol Cell 98:183–193

    PubMed  CAS  Google Scholar 

  65. Medley QG, Buchbinder EG, Tachibana K, Ngo H, Serra-Pages C, Streuli M (2003) Signaling between focal adhesion kinase and trio. J Biol Chem 278:13265–13270

    PubMed  CAS  Google Scholar 

  66. Medley QG, Serra-Pages C, Iannotti E et al (2000) The trio guanine nucleotide exchange factor is a RhoA target. Binding of RhoA to the trio immunoglobulin-like domain. J Biol Chem 275:36116–36123

    PubMed  CAS  Google Scholar 

  67. Ma XM, Huang JP, Eipper BA, Mains RE (2005) Expression of Trio, a member of the Dbl family of Rho GEFs in the developing rat brain. J Comp Neurol 482:333–348

    PubMed  CAS  Google Scholar 

  68. McPherson CE, Eipper BA, Mains RE (2002) Genomic organization and differential expression of Kalirin isoforms. Gene 284:41–51

    PubMed  CAS  Google Scholar 

  69. Lane J, Martin TA, Mansel RE, Jiang WG (2008) The expression and prognostic value of the guanine nucleotide exchange factors (GEFs) Trio, Vav1 and TIAM-1 in human breast cancer. Int Semin Surg Oncol 5:23

    PubMed  Google Scholar 

  70. Yoshizuka N, Moriuchi R, Mori T et al (2004) 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

    PubMed  CAS  Google Scholar 

  71. Blangy A, Bouquier N, Gauthier-Rouviere C et al (2006) Identification of TRIO-GEFD1 chemical inhibitors using the yeast exchange assay. Biol Cell 98:511–522

    PubMed  CAS  Google Scholar 

  72. Bouquier N, Fromont S, Zeeh JC et al (2009) Aptamer-derived peptides as potent inhibitors of the oncogenic RhoGEF Tgat. Chem Biol 16:391–400

    PubMed  CAS  Google Scholar 

  73. Bouquier N, Vignal E, Charrasse S et al (2009) A cell active chemical GEF inhibitor selectively targets the Trio/RhoG/Rac1 signaling pathway. Chem Biol 16:657–666

    PubMed  CAS  Google Scholar 

  74. Gao Y, Dickerson JB, Guo F, Zheng J, Zheng Y (2004) Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc Natl Acad Sci U S A 101:7618–7623

    PubMed  CAS  Google Scholar 

  75. Shinohara M, Terada Y, Iwamatsu A et al (2002) SWAP-70 is a guanine-nucleotide-exchange factor that mediates signalling of membrane ruffling. Nature 416:759–763

    PubMed  CAS  Google Scholar 

  76. Hilpela P, Oberbanscheidt P, Hahne P et al (2003) SWAP-70 identifies a transitional subset of actin filaments in motile cells. Mol Biol Cell 14:3242–3253

    PubMed  Google Scholar 

  77. Murugan AK, Ihara S, Tokuda E, Uematsu K, Tsuchida N, Fukui Y (2008) SWAP-70 is important for invasive phenotypes of mouse embryo fibroblasts transformed by v-Src. IUBMB Life 60:236–240

    PubMed  CAS  Google Scholar 

  78. Miki T (2000) Malignant transformation and regulation of cell division by a Rho exchange factor ECT2. Seikagaku 72:1249–1253

    PubMed  CAS  Google Scholar 

  79. Miki T, Smith CL, Long JE, Eva A, Fleming TP (1993) Oncogene ect2 is related to regulators of small GTP-binding proteins. Nature 362:462–465

    PubMed  CAS  Google Scholar 

  80. Saito S, Liu XF, Kamijo K et al (2004) Deregulation and mislocalization of the cytokinesis regulator ECT2 activate the Rho signaling pathways leading to malignant transformation. J Biol Chem 279:7169–7179

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  83. Roversi G, Pfundt R, Moroni RF et al (2006) Identification of novel genomic markers related to progression to glioblastoma through genomic profiling of 25 primary glioma cell lines. Oncogene 25:1571–1583

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  85. Saito S, Tatsumoto T, Lorenzi MV et al (2003) 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

    PubMed  CAS  Google Scholar 

  86. Hornstein I, Alcover A, Katzav S (2004) Vav proteins, masters of the world of cytoskeleton organization. Cell Signal 16:1–11

    PubMed  CAS  Google Scholar 

  87. Bustelo XR (2000) Regulatory and signaling properties of the Vav family. Mol Cell Biol 20:1461–1477

    PubMed  CAS  Google Scholar 

  88. Dong Z, Liu Y, Lu S et al (2006) Vav3 oncogene is overexpressed and regulates cell growth and androgen receptor activity in human prostate cancer. Mol Endocrinol 20:2315–2325

    PubMed  CAS  Google Scholar 

  89. Lee K, Liu Y, Mo JQ, Zhang J, Dong Z, Lu S (2008) Vav3 oncogene activates estrogen ­receptor and its overexpression may be involved in human breast cancer. BMC Cancer 8:158

    PubMed  Google Scholar 

  90. Colomba A, Courilleau D, Ramel D et al (2008) Activation of Rac1 and the exchange factor Vav3 are involved in NPM-ALK signaling in anaplastic large cell lymphomas. Oncogene 27:2728–2736

    PubMed  CAS  Google Scholar 

  91. Hunter SG, Zhuang G, Brantley-Sieders D, Swat W, Cowan CW, Chen J (2006) Essential role of Vav family guanine nucleotide exchange factors in EphA receptor-mediated angiogenesis. Mol Cell Biol 26:4830–4842

    PubMed  CAS  Google Scholar 

  92. Miyamoto Y, Yamauchi J (2010) Cellular signaling of Dock family proteins in neural ­function. Cell Signal 22:175–182

    PubMed  CAS  Google Scholar 

  93. Nishihara H, Kobayashi S, Hashimoto Y et al (1999) Non-adherent cell-specific expression of DOCK2, a member of the human CDM-family proteins. Biochim Biophys Acta 1452:179–187

    PubMed  CAS  Google Scholar 

  94. Fukui Y, Hashimoto O, Sanui T et al (2001) Haematopoietic cell-specific CDM family protein DOCK2 is essential for lymphocyte migration. Nature 412:826–831

    PubMed  CAS  Google Scholar 

  95. Nombela-Arrieta C, Mempel TR, Soriano SF et al (2007) A central role for DOCK2 during interstitial lymphocyte motility and sphingosine-1-phosphate-mediated egress. J Exp Med 204:497–510

    PubMed  CAS  Google Scholar 

  96. Reif K, Cyster J (2002) The CDM protein DOCK2 in lymphocyte migration. Trends Cell Biol 12:368–373

    PubMed  CAS  Google Scholar 

  97. Nishihara H, Maeda M, Oda A et al (2002) DOCK2 associates with CrkL and regulates Rac1 in human leukemia cell lines. Blood 100:3968–3974

    PubMed  CAS  Google Scholar 

  98. Sanui T, Inayoshi A, Noda M et al (2003) DOCK2 regulates Rac activation and cytoskeletal reorganization through interaction with ELMO1. Blood 102:2948–2950

    PubMed  CAS  Google Scholar 

  99. Janardhan A, Swigut T, Hill B, Myers MP, Skowronski J (2004) HIV-1 Nef binds the DOCK2-ELMO1 complex to activate rac and inhibit lymphocyte chemotaxis. PLoS Biol 2:E6

    PubMed  Google Scholar 

  100. Shulman Z, Pasvolsky R, Woolf E et al (2006) DOCK2 regulates chemokine-triggered lateral lymphocyte motility but not transendothelial migration. Blood 108:2150–2158

    PubMed  CAS  Google Scholar 

  101. Garcia-Bernal D, Sotillo-Mallo E, Nombela-Arrieta C et al (2006) DOCK2 is required for chemokine-promoted human T lymphocyte adhesion under shear stress mediated by the ­integrin alpha4beta1. J Immunol 177:5215–5225

    PubMed  CAS  Google Scholar 

  102. Kunisaki Y, Nishikimi A, Tanaka Y et al (2006) DOCK2 is a Rac activator that regulates motility and polarity during neutrophil chemotaxis. J Cell Biol 174:647–652

    PubMed  CAS  Google Scholar 

  103. Sai J, Raman D, Liu Y, Wikswo J, Richmond A (2008) Parallel phosphatidylinositol 3-kinase (PI3K)-dependent and Src-dependent pathways lead to CXCL8-mediated Rac2 activation and chemotaxis. J Biol Chem 283:26538–26547

    PubMed  CAS  Google Scholar 

  104. Nishikimi A, Fukuhara H, Su W et al (2009) Sequential regulation of DOCK2 dynamics by two phospholipids during neutrophil chemotaxis. Science 324:384–387

    PubMed  CAS  Google Scholar 

  105. Wang L, Nishihara H, Kimura T et al (2010) DOCK2 regulates cell proliferation through Rac and ERK activation in B cell lymphoma. Biochem Biophys Res Commun 395:111–115

    PubMed  CAS  Google Scholar 

  106. El Haibi CP, Sharma PK, Singh R et al (2010) PI3Kp110-, Src-, FAK-dependent and DOCK2-independent migration and invasion of CXCL13-stimulated prostate cancer cells. Mol Cancer 9:85

    PubMed  Google Scholar 

  107. Gotoh K, Tanaka Y, Nishikimi A et al (2010) Selective control of type I IFN induction by the Rac activator DOCK2 during TLR-mediated plasmacytoid dendritic cell activation. J Exp Med 207:721–730

    PubMed  CAS  Google Scholar 

  108. Yajnik V, Paulding C, Sordella R et al (2003) DOCK4, a GTPase activator, is disrupted during tumorigenesis. Cell 112:673–684

    PubMed  CAS  Google Scholar 

  109. Pagnamenta AT, Bacchelli E, de Jonge MV et al (2010) Characterization of a family with rare deletions in CNTNAP5 and DOCK4 suggests novel risk loci for autism and dyslexia. Biol Psychiatry 68:320–328

    PubMed  CAS  Google Scholar 

  110. Poelmans G, Buitelaar JK, Pauls DL, Franke B (2011) A theoretical molecular network for dyslexia: integrating available genetic findings. Mol Psychiatry 16(4):365–382

    PubMed  CAS  Google Scholar 

  111. Gadd S, Sredni ST, Huang CC, Perlman EJ (2010) Rhabdoid tumor: gene expression clues to pathogenesis and potential therapeutic targets. Lab Invest 90:724–738

    PubMed  CAS  Google Scholar 

  112. Maestrini E, Pagnamenta AT, Lamb JA et al (2010) High-density SNP association study and copy number variation analysis of the AUTS1 and AUTS5 loci implicate the IMMP2L-DOCK4 gene region in autism susceptibility. Mol Psychiatry 15:954–968

    PubMed  CAS  Google Scholar 

  113. Kuo KT, Guan B, Feng Y et al (2009) Analysis of DNA copy number alterations in ovarian serous tumors identifies new molecular genetic changes in low-grade and high-grade ­carcinomas. Cancer Res 69:4036–4042

    PubMed  CAS  Google Scholar 

  114. Liang H, Castro PD, Ma J, Nagarajan L (2005) Finer delineation and transcript map of the 7q31 locus deleted in myeloid neoplasms. Cancer Genet Cytogenet 162:151–159

    PubMed  CAS  Google Scholar 

  115. Hiramoto K, Negishi M, Katoh H (2006) Dock4 is regulated by RhoG and promotes Rac-dependent cell migration. Exp Cell Res 312:4205–4216

    PubMed  CAS  Google Scholar 

  116. Ueda S, Fujimoto S, Hiramoto K, Negishi M, Katoh H (2008) Dock4 regulates dendritic development in hippocampal neurons. J Neurosci Res 86:3052–3061

    PubMed  CAS  Google Scholar 

  117. Pannekoek WJ, Kooistra MR, Zwartkruis FJ, Bos JL (2009) Cell-cell junction formation: the role of Rap1 and Rap1 guanine nucleotide exchange factors. Biochim Biophys Acta 1788:790–796

    PubMed  CAS  Google Scholar 

  118. Hiramoto-Yamaki N, Takeuchi S, Ueda S et al (2010) Ephexin4 and EphA2 mediate cell migration through a RhoG-dependent mechanism. J Cell Biol 190:461–477

    PubMed  CAS  Google Scholar 

  119. Upadhyay G, Goessling W, North TE, Xavier R, Zon LI, Yajnik V (2008) Molecular association between beta-catenin degradation complex and Rac guanine exchange factor DOCK4 is essential for Wnt/beta-catenin signaling. Oncogene 27:5845–5855

    PubMed  CAS  Google Scholar 

  120. Watabe-Uchida M, John KA, Janas JA, Newey SE, Van Aelst L (2006) The Rac activator DOCK7 regulates neuronal polarity through local phosphorylation of stathmin/Op18. Neuron 51:727–739

    PubMed  CAS  Google Scholar 

  121. Pinheiro EM, Gertler FB (2006) Nervous Rac: DOCK7 regulation of axon formation. Neuron 51:674–676

    PubMed  CAS  Google Scholar 

  122. Yamauchi J, Miyamoto Y, Chan JR, Tanoue A (2008) ErbB2 directly activates the exchange factor Dock7 to promote Schwann cell migration. J Cell Biol 181:351–365

    PubMed  CAS  Google Scholar 

  123. Guo L, Ying W, Zhang J et al (2010) Tandem affinity purification and identification of the human TSC1 protein complex. Acta Biochim Biophys Sin (Shanghai) 42:266–273

    CAS  Google Scholar 

  124. Rosner M, Hanneder M, Siegel N, Valli A, Hengstschlager M (2008) The tuberous sclerosis gene products hamartin and tuberin are multifunctional proteins with a wide spectrum of interacting partners. Mutat Res 658:234–246

    PubMed  CAS  Google Scholar 

  125. Nellist M, Burgers PC, van den Ouweland AM, Halley DJ, Luider TM (2005) Phosphorylation and binding partner analysis of the TSC1-TSC2 complex. Biochem Biophys Res Commun 333:818–826

    PubMed  CAS  Google Scholar 

  126. Meller N, Irani-Tehrani M, Kiosses WB, Del Pozo MA, Schwartz MA (2002) Zizimin1, a novel Cdc42 activator, reveals a new GEF domain for Rho proteins. Nat Cell Biol 4:639–647

    PubMed  CAS  Google Scholar 

  127. Meller N, Westbrook MJ, Shannon JD, Guda C, Schwartz MA (2008) Function of the N-terminus of zizimin1: autoinhibition and membrane targeting. Biochem J 409:525–533

    PubMed  CAS  Google Scholar 

  128. Kuramoto K, Negishi M, Katoh H (2009) Regulation of dendrite growth by the Cdc42 ­activator Zizimin1/Dock9 in hippocampal neurons. J Neurosci Res 87:1794–1805

    PubMed  CAS  Google Scholar 

  129. Kwofie MA, Skowronski J (2008) Specific recognition of Rac2 and Cdc42 by DOCK2 and DOCK9 guanine nucleotide exchange factors. J Biol Chem 283:3088–3096

    PubMed  CAS  Google Scholar 

  130. Yang J, Zhang Z, Roe SM, Marshall CJ, Barford D (2009) Activation of Rho GTPases by DOCK exchange factors is mediated by a nucleotide sensor. Science 325:1398–1402

    PubMed  CAS  Google Scholar 

  131. Rittinger K (2009) Snapshots form a big picture of guanine nucleotide exchange. Sci Signal 2:63

    Google Scholar 

  132. Hasegawa H, Kiyokawa E, Tanaka S et al (1996) DOCK180, a major CRK-binding protein, alters cell morphology upon translocation to the cell membrane. Mol Cell Biol 16:1770–1776

    PubMed  CAS  Google Scholar 

  133. Matsuda M, Ota S, Tanimura R et al (1996) Interaction between the amino-terminal SH3 domain of CRK and its natural target proteins. J Biol Chem 271:14468–14472

    PubMed  CAS  Google Scholar 

  134. Kiyokawa E, Hashimoto Y, Kobayashi S, Sugimura H, Kurata T, Matsuda M (1998) Activation of Rac1 by a Crk SH3-binding protein, DOCK180. Genes Dev 12:3331–3336

    PubMed  CAS  Google Scholar 

  135. Kiyokawa E, Hashimoto Y, Kurata T, Sugimura H, Matsuda M (1998) Evidence that DOCK180 up-regulates signals from the CrkII-p130(Cas) complex. J Biol Chem 273:24479–24484

    PubMed  CAS  Google Scholar 

  136. Cheresh DA, Leng J, Klemke RL (1999) Regulation of cell contraction and membrane ­ruffling by distinct signals in migratory cells. J Cell Biol 146:1107–1116

    PubMed  CAS  Google Scholar 

  137. Erickson MR, Galletta BJ, Abmayr SM (1997) Drosophila myoblast city encodes a ­conserved protein that is essential for myoblast fusion, dorsal closure, and cytoskeletal organization. J Cell Biol 138:589–603

    PubMed  CAS  Google Scholar 

  138. Nolan KM, Barrett K, Lu Y, Hu KQ, Vincent S, Settleman J (1998) Myoblast city, the Drosophila homolog of DOCK180/CED-5, is required in a Rac signaling pathway utilized for multiple developmental processes. Genes Dev 12:3337–3342

    PubMed  CAS  Google Scholar 

  139. Wu YC, Horvitz HR (1998) C. elegans phagocytosis and cell-migration protein CED-5 is similar to human DOCK180. Nature 392:501–504

    PubMed  CAS  Google Scholar 

  140. Reddien PW, Horvitz HR (2000) CED-2/CrkII and CED-10/Rac control phagocytosis and cell migration in Caenorhabditis elegans. Nat Cell Biol 2:131–136

    PubMed  CAS  Google Scholar 

  141. Tosello-Trampont A-C, Brugnera E, Ravichandran KS (2001) Evidence for a Conserved Role for CrkII and Rac in Engulfment of Apoptotic Cells. J Biol Chem 276:13797–13802

    PubMed  CAS  Google Scholar 

  142. Zhou Z, Caron E, Hartwieg E, Hall A, Horvitz HR (2001) The C. elegans PH domain protein CED-12 regulates cytoskeletal reorganization via a Rho/Rac GTPase signaling pathway. Dev Cell 1:477–489

    PubMed  CAS  Google Scholar 

  143. Wang X, Wu YC, Fadok VA et al (2003) Cell corpse engulfment mediated by C. elegans phosphatidylserine receptor through CED-5 and CED-12. Science 302:1563–1566

    PubMed  CAS  Google Scholar 

  144. Valles AM, Beuvin M, Boyer B (2004) Activation of Rac1 by paxillin-Crk-DOCK180 signaling complex is antagonized by Rap1 in migrating NBT-II cells. J Biol Chem 279:44490–44496

    PubMed  CAS  Google Scholar 

  145. Lu M, Kinchen JM, Rossman KL et al (2005) A Steric-inhibition model for regulation of nucleotide exchange via the Dock180 family of GEFs. Curr Biol 15:371–377

    PubMed  CAS  Google Scholar 

  146. Moore CA, Parkin CA, Bidet Y, Ingham PW (2007) A role for the Myoblast city homologues Dock1 and Dock5 and the adaptor proteins Crk and Crk-like in zebrafish myoblast fusion. Development 134:3145–3153

    PubMed  CAS  Google Scholar 

  147. Pajcini KV, Pomerantz JH, Alkan O, Doyonnas R, Blau HM (2008) Myoblasts and ­macrophages share molecular components that contribute to cell-cell fusion. J Cell Biol 180:1005–1019

    PubMed  CAS  Google Scholar 

  148. Kobayashi S, Shirai T, Kiyokawa E, Mochizuki N, Matsuda M, Fukui Y (2001) Membrane recruitment of DOCK180 by binding to PtdIns(3,4,5)P3. Biochem J 354:73–78

    PubMed  CAS  Google Scholar 

  149. Cote JF, Vuori K (2006) In vitro guanine nucleotide exchange activity of DHR-2/DOCKER/CZH2 domains. Methods Enzymol 406:41–57

    PubMed  CAS  Google Scholar 

  150. Gumienny TL, Brugnera E, Tosello-Trampont AC et al (2001) CED-12/ELMO, a novel member of the CrkII/Dock180/Rac pathway, is required for phagocytosis and cell migration. Cell 107:27–41

    PubMed  CAS  Google Scholar 

  151. Wu YC, Tsai MC, Cheng LC, Chou CJ, Weng NY (2001) C. elegans CED-12 acts in the conserved crkII/DOCK180/Rac pathway to control cell migration and cell corpse engulfment. Dev Cell 1:491–502

    PubMed  CAS  Google Scholar 

  152. Grimsley CM, Kinchen JM, Tosello-Trampont AC et al (2004) Dock180 and ELMO1 ­proteins cooperate to promote evolutionarily conserved Rac-dependent cell migration. J Biol Chem 279:6087–6097

    PubMed  CAS  Google Scholar 

  153. Geisbrecht ER, Haralalka S, Swanson SK, Florens L, Washburn MP, Abmayr SM (2008) Drosophila ELMO/CED-12 interacts with Myoblast city to direct myoblast fusion and ommatidial organization. Dev Biol 314:137–149

    PubMed  CAS  Google Scholar 

  154. Akakura S, Kar B, Singh S et al (2005) C-terminal SH3 domain of CrkII regulates the ­assembly and function of the DOCK180/ELMO Rac-GEF. J Cell Physiol 204:344–351

    PubMed  CAS  Google Scholar 

  155. Zaidel-Bar R, Kam Z, Geiger B (2005) Polarized downregulation of the paxillin-p130CAS-Rac1 pathway induced by shear flow. J Cell Sci 118:3997–4007

    PubMed  CAS  Google Scholar 

  156. Tosello-Trampont AC, Kinchen JM, Brugnera E, Haney LB, Hengartner MO, Ravichandran KS (2007) Identification of two signaling submodules within the CrkII/ELMO/Dock180 pathway regulating engulfment of apoptotic cells. Cell Death Differ 14:963–972

    PubMed  CAS  Google Scholar 

  157. Handa Y, Suzuki M, Ohya K et al (2007) Shigella IpgB1 promotes bacterial entry through the ELMO-Dock180 machinery. Nat Cell Biol 9:121–128

    PubMed  CAS  Google Scholar 

  158. Meller J, Vidali L, Schwartz MA (2008) Endogenous RhoG is dispensable for integrin-­mediated cell spreading but contributes to Rac-independent migration. J Cell Sci 121:1981–1989

    PubMed  CAS  Google Scholar 

  159. Para A, Krischke M, Merlot S et al (2009) Dictyostelium Dock180-related RacGEFs regulate the actin cytoskeleton during cell motility. Mol Biol Cell 20:699–707

    PubMed  CAS  Google Scholar 

  160. Makino Y, Tsuda M, Ichihara S et al (2006) Elmo1 inhibits ubiquitylation of Dock180. J Cell Sci 119:923–932

    PubMed  CAS  Google Scholar 

  161. Cho NK, Keyes L, Johnson E et al (2002) Developmental control of blood cell migration by the Drosophila VEGF pathway. Cell 108:865–876

    PubMed  CAS  Google Scholar 

  162. Kraut R, Menon K, Zinn K (2001) A gain-of-function screen for genes controlling motor axon guidance and synaptogenesis in Drosophila. Curr Biol 11:417–430

    PubMed  CAS  Google Scholar 

  163. Duchek P, Somogyi K, Jekely G, Beccari S, Rorth P (2001) Guidance of cell migration by the Drosophila PDGF/VEGF receptor. Cell 107:17–26

    PubMed  CAS  Google Scholar 

  164. Fulga TA, Rorth P (2002) Invasive cell migration is initiated by guided growth of long cellular extensions. Nat Cell Biol 4:715–719

    PubMed  CAS  Google Scholar 

  165. Ishimaru S, Ueda R, Hinohara Y, Ohtani M, Hanafusa H (2004) PVR plays a critical role via JNK activation in thorax closure during Drosophila metamorphosis. EMBO J 23:3984–3994

    PubMed  CAS  Google Scholar 

  166. Bianco A, Poukkula M, Cliffe A et al (2007) Two distinct modes of guidance signalling ­during collective migration of border cells. Nature 448:362–365

    PubMed  CAS  Google Scholar 

  167. Albert ML, Kim JI, Birge RB (2000) alphavbeta5 integrin recruits the CrkII-Dock180-rac1 complex for phagocytosis of apoptotic cells. Nat Cell Biol 2:899–905

    PubMed  CAS  Google Scholar 

  168. Gu J, Sumida Y, Sanzen N, Sekiguchi K (2001) Laminin-10/11 and fibronectin differentially regulate integrin-dependent Rho and Rac activation via p130(Cas)-CrkII-DOCK180 ­pathway. J Biol Chem 276:27090–27097

    PubMed  CAS  Google Scholar 

  169. Gustavsson A, Yuan M, Fallman M (2004) Temporal dissection of beta1-integrin signaling indicates a role for p130Cas-Crk in filopodia formation. J Biol Chem 279:22893–22901

    PubMed  CAS  Google Scholar 

  170. Smith HW, Marra P, Marshall CJ (2008) uPAR promotes formation of the p130Cas-Crk ­complex to activate Rac through DOCK180. J Cell Biol 182:777–790

    PubMed  CAS  Google Scholar 

  171. Lee WL, Cosio G, Ireton K, Grinstein S (2007) Role of CrkII in Fc{gamma} Receptor-mediated Phagocytosis. J Biol Chem 282:11135–11143

    PubMed  CAS  Google Scholar 

  172. Payne SL, Hendrix MJ, Kirschmann DA (2006) Lysyl oxidase regulates actin filament formation through the p130(Cas)/Crk/DOCK180 signaling complex. J Cell Biochem 98:827–837

    PubMed  CAS  Google Scholar 

  173. Laurin M, Fradet N, Blangy A, Hall A, Vuori K, Cote JF (2008) The atypical Rac activator Dock180 (Dock1) regulates myoblast fusion in vivo. Proc Natl Acad Sci U S A 105(40):15446–15451

    PubMed  CAS  Google Scholar 

  174. Epting D, Wendik B, Bennewitz K, Dietz CT, Driever W, Kroll J (2010) The Rac1 regulator ELMO1 controls vascular morphogenesis in zebrafish. Circ Res 107:45–55

    PubMed  CAS  Google Scholar 

  175. Park D, Tosello-Trampont AC, Elliott MR et al (2007) BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450:430–434

    PubMed  CAS  Google Scholar 

  176. Elliott MR, Zheng S, Park D et al (2010) Unexpected requirement for ELMO1 in clearance of apoptotic germ cells in vivo. Nature 467:333–337

    PubMed  CAS  Google Scholar 

  177. Elliott MR, Ravichandran KS (2010) ELMO1 signaling in apoptotic germ cell clearance and spermatogenesis. Ann N Y Acad Sci 1209:30–36

    PubMed  CAS  Google Scholar 

  178. Shimazaki A, Kawamura Y, Kanazawa A et al (2005) Genetic variations in the gene encoding ELMO1 are associated with susceptibility to diabetic nephropathy. Diabetes 54:1171–1178

    PubMed  CAS  Google Scholar 

  179. Pezzolesi MG, Katavetin P, Kure M et al (2009) Confirmation of genetic associations at ELMO1 in the GoKinD collection supports its role as a susceptibility gene in diabetic nephropathy. Diabetes 58:2698–2702

    PubMed  CAS  Google Scholar 

  180. Leak TS, Perlegas PS, Smith SG et al (2009) Variants in intron 13 of the ELMO1 gene are associated with diabetic nephropathy in African Americans. Ann Hum Genet 73:152–159

    PubMed  CAS  Google Scholar 

  181. Doria A (2010) Genetics of diabetes complications. Curr Diab Rep 10:467–475

    PubMed  CAS  Google Scholar 

  182. Phillips HS, Kharbanda S, Chen R et al (2006) Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9:157–173

    PubMed  CAS  Google Scholar 

  183. Hsia DA, Mitra SK, Hauck CR et al (2003) Differential regulation of cell motility and invasion by FAK. J Cell Biol 160:753–767

    PubMed  CAS  Google Scholar 

  184. Wang H, Linghu H, Wang J et al (2010) The role of Crk/Dock180/Rac1 pathway in the malignant behavior of human ovarian cancer cell SKOV3. Tumour Biol 31:59–67

    PubMed  Google Scholar 

  185. Jarzynka MJ, Hu B, Hui KM et al (2007) ELMO1 and Dock180, a bipartite Rac1 guanine nucleotide exchange factor, promote human glioma cell invasion. Cancer Res 67:7203–7211

    PubMed  CAS  Google Scholar 

  186. Foster D, Parrish-Novak J, Fox B, Xu W (2004) Cytokine-receptor pairing: accelerating discovery of cytokine function. Nat Rev Drug Discov 3:160–170

    PubMed  CAS  Google Scholar 

  187. Locksley RM, Killeen N, Lenardo MJ (2001) The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104:487–501

    PubMed  CAS  Google Scholar 

  188. Baker SJ, Reddy EP (1998) Modulation of life and death by the TNF receptor superfamily. Oncogene 17:3261–3270

    PubMed  Google Scholar 

  189. Fesik SW (2000) Insights into programmed cell death through structural biology. Cell 103:273–282

    PubMed  CAS  Google Scholar 

  190. Brown SA, Richards CM, Hanscom HN, Feng SL, Winkles JA (2003) The Fn14 cytoplasmic tail binds tumour-necrosis-factor-receptor-associated factors 1, 2, 3 and 5 and mediates nuclear factor-kappaB activation. Biochem J 371:395–403

    PubMed  CAS  Google Scholar 

  191. Willis AL, Tran NL, Chatigny JM et al (2008) The fibroblast growth factor-inducible 14 receptor is highly expressed in HER2-positive breast tumors and regulates breast cancer cell invasive capacity. Mol Cancer Res 6:725–734

    PubMed  CAS  Google Scholar 

  192. Meighan-Mantha RL, Hsu DK, Guo Y et al (1999) The mitogen-inducible Fn14 gene encodes a type I transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem 274:33166–33176

    PubMed  CAS  Google Scholar 

  193. Wiley SR, Winkles JA (2003) TWEAK, a member of the TNF superfamily, is a ­multifunctional cytokine that binds the TweakR/Fn14 receptor. Cytokine Growth Factor Rev 14:241–249

    PubMed  CAS  Google Scholar 

  194. Feng SL, Guo Y, Factor VM et al (2000) The Fn14 immediate-early response gene is induced during liver regeneration and highly expressed in both human and murine hepatocellular carcinomas. Am J Pathol 156:1253–1261

    PubMed  CAS  Google Scholar 

  195. Wiley SR, Cassiano L, Lofton T et al (2001) A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis. Immunity 15:837–846

    PubMed  CAS  Google Scholar 

  196. Tran NL, McDonough WS, Donohue PJ et al (2003) The human Fn14 receptor gene is up-regulated in migrating glioma cells in vitro and overexpressed in advanced glial tumors. Am J Pathol 162:1313–1321

    PubMed  CAS  Google Scholar 

  197. Tran NL, McDonough WS, Savitch BA, Sawyer TF, Winkles JA, Berens ME (2005) The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-fibroblast growth factor-inducible 14 (Fn14) signaling system regulates glioma cell survival via NFkappaB pathway activation and BCL-XL/BCL-W expression. J Biol Chem 280:3483–3492

    PubMed  CAS  Google Scholar 

  198. Han S, Yoon K, Lee K et al (2003) TNF-related weak inducer of apoptosis receptor, a TNF receptor superfamily member, activates NF-kappa B through TNF receptor-associated factors. Biochem Biophys Res Commun 305:789–796

    PubMed  CAS  Google Scholar 

  199. Tanabe K, Bonilla I, Winkles JA, Strittmatter SM (2003) Fibroblast growth factor-­inducible-14 is induced in axotomized neurons and promotes neurite outgrowth. J Neurosci 23:9675–9686

    PubMed  CAS  Google Scholar 

  200. Watts GS, Tran NL, Berens ME et al (2007) Identification of Fn14/TWEAK receptor as a potential therapeutic target in esophageal adenocarcinoma. Int J Cancer 121:2132–2139

    PubMed  CAS  Google Scholar 

  201. Hu S, Tamada K, Ni J, Vincenz C, Chen L (1999) Characterization of TNFRSF19, a novel member of the tumor necrosis factor receptor superfamily. Genomics 62:103–107

    PubMed  CAS  Google Scholar 

  202. Park JB, Yiu G, Kaneko S et al (2005) A TNF receptor family member, TROY, is a coreceptor with Nogo receptor in mediating the inhibitory activity of myelin inhibitors. Neuron 45:345–351

    PubMed  CAS  Google Scholar 

  203. Pispa J, Mikkola ML, Mustonen T, Thesleff I (2003) Ectodysplasin, Edar and TNFRSF19 are expressed in complementary and overlapping patterns during mouse embryogenesis. Gene Expr Patterns 3:675–679

    PubMed  CAS  Google Scholar 

  204. Hisaoka T, Morikawa Y, Kitamura T, Senba E (2004) Expression of a member of tumor necrosis factor receptor superfamily, TROY, in the developing olfactory system. Glia 45:313–324

    PubMed  Google Scholar 

  205. Shao Z, Browning JL, Lee X et al (2005) TAJ/TROY, an orphan TNF receptor family member, binds Nogo-66 receptor 1 and regulates axonal regeneration. Neuron 45:353–359

    PubMed  CAS  Google Scholar 

  206. Hisaoka T, Morikawa Y, Kitamura T, Senba E (2003) Expression of a member of tumor necrosis factor receptor superfamily, TROY, in the developing mouse brain. Brain Res Dev Brain Res 143:105–109

    PubMed  CAS  Google Scholar 

  207. Paulino VM, Yang Z, Kloss JM et al (2010) TROY (TNFRSF19) is overexpressed in advanced glial tumors and promotes glioblastoma cell invasion via Pyk2-Rac1 signaling. Mol Cancer Res 8(11):1558–1567

    PubMed  CAS  Google Scholar 

  208. Girault JA, Costa A, Derkinderen P, Studler JM, Toutant M (1999) FAK and PYK2/CAKbeta in the nervous system: a link between neuronal activity, plasticity and survival? Trends Neurosci 22:257–263

    PubMed  CAS  Google Scholar 

  209. Avraham H, Park SY, Schinkmann K, Avraham S (2000) RAFTK/Pyk2-mediated cellular signalling. Cell Signal 12:123–133

    PubMed  CAS  Google Scholar 

  210. Sun CK, Ng KT, Sun BS et al (2007) The significance of proline-rich tyrosine kinase2 (Pyk2) on hepatocellular carcinoma progression and recurrence. Br J Cancer 97:50–57

    PubMed  CAS  Google Scholar 

  211. Sun CK, Man K, Ng KT et al (2008) Proline-rich tyrosine kinase 2 (Pyk2) promotes proliferation and invasiveness of hepatocellular carcinoma cells through c-Src/ERK activation. Carcinogenesis 29:2096–2105

    PubMed  CAS  Google Scholar 

  212. Behmoaram E, Bijian K, Jie S et al (2008) Focal adhesion kinase-related proline-rich tyrosine kinase 2 and focal adhesion kinase are co-overexpressed in early-stage and invasive ErbB-2-positive breast cancer and cooperate for breast cancer cell tumorigenesis and ­invasiveness. Am J Pathol 173:1540–1550

    PubMed  CAS  Google Scholar 

  213. Gutenberg A, Bruck W, Buchfelder M, Ludwig HC (2004) Expression of tyrosine kinases FAK and Pyk2 in 331 human astrocytomas. Acta Neuropathol 108:224–230

    PubMed  CAS  Google Scholar 

  214. Mariani L, Beaudry C, McDonough WS et al (2001) Glioma cell motility is associated with reduced transcription of proapoptotic and proliferation genes: a cDNA microarray analysis. J Neurooncol 53:161–176

    PubMed  CAS  Google Scholar 

  215. Hoelzinger DB, Mariani L, Weis J et al (2005) Gene expression profile of glioblastoma ­multiforme invasive phenotype points to new therapeutic targets. Neoplasia 7:7–16

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shi-Yuan Cheng .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2011 Springer Science+Business Media B.V.

About this chapter

Cite this chapter

Hu, B. et al. (2011). Rho GTPases and Their Activators, Guanine Nucleotide Exchange Factors (GEFs): Their Roles in Glioma Cell Invasion. In: Fatatis, A. (eds) Signaling Pathways and Molecular Mediators in Metastasis. Springer, Dordrecht. https://doi.org/10.1007/978-94-007-2558-4_6

Download citation

Publish with us

Policies and ethics