4,210
Views
52
CrossRef citations to date
0
Altmetric
Reviews

The RhoB small GTPase in physiology and disease

ORCID Icon & ORCID Icon
Pages 384-393 | Received 28 Sep 2016, Accepted 23 Oct 2016, Published online: 22 Nov 2016

References

  • Thumkeo D, Watanabe S, Narumiya S. Physiological roles of Rho and Rho effectors in mammals. Eur J Cell Biol 2013; 92(10-11):303-15; PMID:24183240; https://doi.org/10.1016/j.ejcb.2013.09.002
  • Ridley AJ. Rho GTPase signalling in cell migration. Curr Opin Cell Biol 2015; 36:103-12; PMID:26363959; https://doi.org/10.1016/j.ceb.2015.08.005
  • Kersey PJ, Allen JE, Armean I, Boddu S, Bolt BJ, Carvalho-Silva D, Christensen M, Davis P, Falin LJ, Grabmueller C, et al. Ensembl Genomes 2016: more genomes, more complexity. Nucleic Acids Res 2016; 44(D1):D574-80; PMID:26578574; https://doi.org/10.1093/nar/gkv1209
  • Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr Protoc Bioinformatics 2016; 54:1
  • Schaefer A, Reinhard NR, Hordijk PL. Toward understanding RhoGTPase specificity: structure, function and local activation. Small GTPases 2014; 5(2):6; https://doi.org/10.4161/21541248.2014.968004
  • Adamson P, Paterson HF, Hall A. Intracellular localization of the P21rho proteins. J Cell Biol 1992; 119(3):617-27; PMID:1383236; https://doi.org/10.1083/jcb.119.3.617
  • Wherlock M, Gampel A, Futter C, Mellor H. Farnesyltransferase inhibitors disrupt EGF receptor traffic through modulation of the RhoB GTPase. J Cell Sci 2004; 117(Pt 15):3221-31; PMID:15226397; https://doi.org/10.1242/jcs.01193
  • Gerald D, Adini I, Shechter S, Perruzzi C, Varnau J, Hopkins B, Kazerounian S, Kurschat P, Blachon S, Khedkar S, et al. RhoB controls coordination of adult angiogenesis and lymphangiogenesis following injury by regulating VEZF1-mediated transcription. Nat Commun 2013; 4:2824; PMID:24280686; https://doi.org/10.1038/ncomms3824
  • Arthur WT, Ellerbroek SM, Der CJ, Burridge K, Wennerberg K. XPLN, a guanine nucleotide exchange factor for RhoA and RhoB, but not RhoC. J Biol Chem 2002; 277(45):42964-72; PMID:12221096; https://doi.org/10.1074/jbc.M207401200
  • Hamel B, Monaghan-Benson E, Rojas RJ, Temple BR, Marston DJ, Burridge K, Sondek J. SmgGDS is a guanine nucleotide exchange factor that specifically activates RhoA and RhoC. J Biol Chem 2011; 286(14):12141-8; PMID:21242305; https://doi.org/10.1074/jbc.M110.191122
  • Cherfils J, Zeghouf M. Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev 2013; 93(1):269-309; PMID:23303910; https://doi.org/10.1152/physrev.00003.2012
  • Zalcman G, Closson V, Camonis J, Honore N, Rousseau-Merck MF, Tavitian A, Olofsson B. RhoGDI-3 is a new GDP dissociation inhibitor (GDI). Identification of a non-cytosolic GDI protein interacting with the small GTP-binding proteins RhoB and RhoG. J Biol Chem 1996; 271(48):30366-74; PMID:8939998; https://doi.org/10.1074/jbc.271.48.30366
  • Wang M, Guo L, Wu Q, Zeng T, Lin Q, Qiao Y, Wang Q, Liu M, Zhang X, Ren L, et al. ATR/Chk1/Smurf1 pathway determines cell fate after DNA damage by controlling RhoB abundance. Nat Commun 2014; 5:4901; PMID:25249323; https://doi.org/10.1038/ncomms5901
  • Xu J, Li L, Yu G, Ying W, Gao Q, Zhang W, Li X, Ding C, Jiang Y, Wei D, et al. The neddylation-cullin 2-RBX1 E3 ligase axis targets tumor suppressor RhoB for degradation in liver cancer. Mol Cell Proteomics 2015; 14(3):499-509; PMID:25540389; https://doi.org/10.1074/mcp.M114.045211
  • Zalcman G, Closson V, Linares-Cruz G, Lerebours F, Honore N, Tavitian A, Olofsson B. Regulation of Ras-related RhoB protein expression during the cell cycle. Oncogene 1995; 10(10):1935-45; PMID:7539118
  • Liu M, Tang Q, Qiu M, Lang N, Li M, Zheng Y, Bi F. miR-21 targets the tumor suppressor RhoB and regulates proliferation, invasion and apoptosis in colorectal cancer cells. FEBS Lett 2011; 585(19):2998-3005; PMID:21872591; https://doi.org/10.1016/j.febslet.2011.08.014
  • Tan Y, Yin H, Zhang H, Fang J, Zheng W, Li D, Li Y, Cao W, Sun C, Liang Y, et al. Sp1-driven up-regulation of miR-19a decreases RHOB and promotes pancreatic cancer. Oncotarget 2015; 6(19):17391-403; PMID:26041879; https://doi.org/10.18632/oncotarget.3975
  • Ho TT, Merajver SD, Lapiere CM, Nusgens BV, Deroanne CF. RhoA-GDP regulates RhoB protein stability. Potential involvement of RhoGDIalpha. J Biol Chem 2008; 283(31):21588-98; PMID:18524772; https://doi.org/10.1074/jbc.M710033200
  • Vega FM, Fruhwirth G, Ng T, Ridley AJ. RhoA and RhoC have distinct roles in migration and invasion by acting through different targets. J Cell Biol 2011; 193(4):655-65; PMID:21576392; https://doi.org/10.1083/jcb.201011038
  • Guilluy C, Garcia-Mata R, Burridge K. Rho protein crosstalk: another social network? Trends Cell Biol 2011; 21(12):718-26; PMID:21924908; https://doi.org/10.1016/j.tcb.2011.08.002
  • Marcos-Ramiro B, Garcia-Weber D, Barroso S, Feito J, Ortega MC, Cernuda-Morollon E, Reglero-Real N, Fernandez-Martin L, Duran MC, Alonso MA, et al. RhoB controls endothelial barrier recovery by inhibiting Rac1 trafficking to the cell border. J Cell Biol 2016; 213(3):385-402; PMID:27138256; https://doi.org/10.1083/jcb.201504038
  • Tillement V, Lajoie-Mazenc I, Casanova A, Froment C, Penary M, Tovar D, Marquez R, Monsarrat B, Favre G, Pradines A. Phosphorylation of RhoB by CK1 impedes actin stress fiber organization and epidermal growth factor receptor stabilization. Exp Cell Res 2008; 314(15):2811-21; PMID:18590726; https://doi.org/10.1016/j.yexcr.2008.06.011
  • Ballif BA, Carey GR, Sunyaev SR, Gygi SP. Large-scale identification and evolution indexing of tyrosine phosphorylation sites from murine brain. J Proteome Res 2008; 7(1):311-8; PMID:18034455; https://doi.org/10.1021/pr0701254
  • Truebestein L, Elsner DJ, Fuchs E, Leonard TA. A molecular ruler regulates cytoskeletal remodelling by the Rho kinases. Nat Commun 2015; 6:10029; PMID:26620183; https://doi.org/10.1038/ncomms10029
  • Hutchinson CL, Lowe PN, McLaughlin SH, Mott HR, Owen D. Differential binding of RhoA, RhoB, and RhoC to protein kinase C-related kinase (PRK) isoforms PRK1, PRK2, and PRK3: PRKs have the highest affinity for RhoB. Biochemistry 2013; 52(45):7999-8011; PMID:24128008; https://doi.org/10.1021/bi401216w
  • Mellor H, Flynn P, Nobes CD, Hall A, Parker PJ. PRK1 is targeted to endosomes by the small GTPase, RhoB. J Biol Chem 1998; 273(9):4811-4; PMID:9478917; https://doi.org/10.1074/jbc.273.9.4811
  • Donnelly SK, Bravo-Cordero JJ, Hodgson L. Rho GTPase isoforms in cell motility: Don't fret, we have FRET. Cell Adh Migr 2014; 8(6):526-34; PMID:25482645; https://doi.org/10.4161/cam.29712
  • Wallar BJ, Deward AD, Resau JH, Alberts AS. RhoB and the mammalian Diaphanous-related formin mDia2 in endosome trafficking. Exp Cell Res 2007; 313(3):560-71; PMID:17198702; https://doi.org/10.1016/j.yexcr.2006.10.033
  • Reinhard NR, van Helden SF, Anthony EC, Yin T, Wu YI, Goedhart J, Gadella TW, Hordijk PL. Spatiotemporal analysis of RhoA/B/C activation in primary human endothelial cells. Sci Rep 2016; 6:25502; PMID:27147504; https://doi.org/10.1038/srep25502
  • Fernandez-Borja M, Janssen L, Verwoerd D, Hordijk P, Neefjes J. RhoB regulates endosome transport by promoting actin assembly on endosomal membranes through Dia1. J Cell Sci 2005; 118(Pt 12):2661-70; PMID:15944396; https://doi.org/10.1242/jcs.02384
  • Adini I, Rabinovitz I, Sun JF, Prendergast GC, Benjamin LE. RhoB controls Akt trafficking and stage-specific survival of endothelial cells during vascular development. Genes Dev 2003; 17(21):2721-32; PMID:14597666; https://doi.org/10.1101/gad.1134603
  • Gampel A, Parker PJ, Mellor H. Regulation of epidermal growth factor receptor traffic by the small GTPase rhoB. Curr Biol 1999; 9(17):955-8; PMID:10508588; https://doi.org/10.1016/S0960-9822(99)80422-9
  • Sandilands E, Cans C, Fincham VJ, Brunton VG, Mellor H, Prendergast GC, Norman JC, Superti-Furga G, Frame MC. RhoB and actin polymerization coordinate Src activation with endosome-mediated delivery to the membrane. Dev Cell 2004; 7(6):855-69; PMID:15572128; https://doi.org/10.1016/j.devcel.2004.09.019
  • Neel NF, Lapierre LA, Goldenring JR, Richmond A. RhoB plays an essential role in CXCR2 sorting decisions. J Cell Sci 2007; 120(Pt 9):1559-71; PMID:17405813; https://doi.org/10.1242/jcs.03437
  • Liu AX, Rane N, Liu JP, Prendergast GC. RhoB is dispensable for mouse development, but it modifies susceptibility to tumor formation as well as cell adhesion and growth factor signaling in transformed cells. Mol Cell Biol 2001; 21(20):6906-12; PMID:11564874; https://doi.org/10.1128/MCB.21.20.6906-6912.2001
  • Bravo-Nuevo A, O'Donnell R, Rosendahl A, Chung JH, Benjamin LE, Odaka C. RhoB deficiency in thymic medullary epithelium leads to early thymic atrophy. Int Immunol 2011; 23(10):593-600; PMID:21865151; https://doi.org/10.1093/intimm/dxr064
  • Henderson DJ, Ybot-Gonzalez P, Copp AJ. RhoB is expressed in migrating neural crest and endocardial cushions of the developing mouse embryo. Mech Dev 2000; 95(1-2):211-4; PMID:10906464; https://doi.org/10.1016/S0925-4773(00)00333-6
  • Santos-Bredariol AS, Belmonte MA, Kihara AH, Santos MF, Hamassaki DE. Small GTP-binding protein RhoB is expressed in glial Muller cells in the vertebrate retina. J Comp Neurol 2006; 494(6):976-85; PMID:16385489; https://doi.org/10.1002/cne.20861
  • McKinnell IW, Makarenkova H, de Curtis I, Turmaine M, Patel K. EphA4, RhoB and the molecular development of feather buds are maintained by the integrity of the actin cytoskeleton. Dev Biol 2004; 270(1):94-105; PMID:15136143; https://doi.org/10.1016/j.ydbio.2004.02.007
  • del Barrio MG, Nieto MA. Overexpression of Snail family members highlights their ability to promote chick neural crest formation. Development 2002; 129(7):1583-93; PMID:11923196
  • Perez-Alcala S, Nieto MA, Barbas JA. LSox5 regulates RhoB expression in the neural tube and promotes generation of the neural crest. Development 2004; 131(18):4455-65; PMID:15306568; https://doi.org/10.1242/dev.01329
  • Vignal E, de Santa Barbara P, Guemar L, Donnay JM, Fort P, Faure S. Expression of RhoB in the developing Xenopus laevis embryo. Gene Expr Patterns 2007; 7(3):282-8; PMID:17049930; https://doi.org/10.1016/j.modgep.2006.09.002
  • Liu JP, Jessell TM. A role for rhoB in the delamination of neural crest cells from the dorsal neural tube. Development 1998; 125(24):5055-67; PMID:9811589
  • Cheung M, Chaboissier MC, Mynett A, Hirst E, Schedl A, Briscoe J. The transcriptional control of trunk neural crest induction, survival, and delamination. Dev Cell 2005; 8(2):179-92; PMID:15691760; https://doi.org/10.1016/j.devcel.2004.12.010
  • Zhang J, Zhu J, Bu X, Cushion M, Kinane TB, Avraham H, Koziel H. Cdc42 and RhoB activation are required for mannose receptor-mediated phagocytosis by human alveolar macrophages. Mol Biol Cell 2005; 16(2):824-34; PMID:15574879; https://doi.org/10.1091/mbc.E04-06-0463
  • Wang XH, Wang Y, Diao F, Lu J. RhoB is involved in lipopolysaccharide-induced inflammation in mouse in vivo and in vitro. J Physiol Biochem 2013; 69(2):189-97; PMID:22869204; https://doi.org/10.1007/s13105-012-0201-z
  • Wheeler AP, Ridley AJ. RhoB affects macrophage adhesion, integrin expression and migration. Exp Cell Res 2007; 313(16):3505-16; PMID:17692842; https://doi.org/10.1016/j.yexcr.2007.07.014
  • Konigs V, Jennings R, Vogl T, Horsthemke M, Bachg AC, Xu Y, Grobe K, Brakebusch C, Schwab A, Bahler M, et al. Mouse macrophages completely lacking Rho subfamily GTPases (RhoA, RhoB, and RhoC) have severe lamellipodial retraction defects, but robust chemotactic navigation and altered motility. J Biol Chem 2014; 289(44):30772-84; PMID:25213860; https://doi.org/10.1074/jbc.M114.563270
  • Huang G, Su J, Zhang M, Jin Y, Wang Y, Zhou P, Lu J. RhoB regulates the function of macrophages in the hypoxia-induced inflammatory response. Cell Mol Immunol 2015; 1-11
  • Skuli N, Monferran S, Delmas C, Lajoie-Mazenc I, Favre G, Toulas C, Cohen-Jonathan-Moyal E. Activation of RhoB by hypoxia controls hypoxia-inducible factor-1alpha stabilization through glycogen synthase kinase-3 in U87 glioblastoma cells. Cancer Res 2006; 66(1):482-9; PMID:16397264; https://doi.org/10.1158/0008-5472.CAN-05-2299
  • Wojciak-Stothard B, Zhao L, Oliver E, Dubois O, Wu Y, Kardassis D, Vasilaki E, Huang M, Mitchell JA, Harrington LS, et al. Role of RhoB in the regulation of pulmonary endothelial and smooth muscle cell responses to hypoxia. Circ Res 2012; 110(11):1423-34; PMID:22539766; https://doi.org/10.1161/CIRCRESAHA.112.264473
  • Rodriguez PL, Sahay S, Olabisi OO, Whitehead IP. ROCK I-mediated activation of NF-kappaB by RhoB. Cell Signall 2007; 19(11):2361-9; PMID:17728102; https://doi.org/10.1016/j.cellsig.2007.07.021
  • Goulidaki N, Alarifi S, Alkahtani SH, Al-Qahtani A, Spandidos DA, Stournaras C, Sourvinos G. RhoB is a component of the human cytomegalovirus assembly complex and is required for efficient viral production. Cell Cycle 2015; 14(17):2748-63; PMID:26114383; https://doi.org/10.1080/15384101.2015.1066535
  • Kroon J, Tol S, van Amstel S, Elias JA, Fernandez-Borja M. The small GTPase RhoB regulates TNFalpha signaling in endothelial cells. PloS One 2013; 8(9):e75031; PMID:24086429; https://doi.org/10.1371/journal.pone.0075031
  • Howe GA, Addison CL. RhoB controls endothelial cell morphogenesis in part via negative regulation of RhoA. Vascular Cell 2012; 4:1; PMID:22316440; https://doi.org/10.1186/2045-824X-4-1
  • Sabatel C, Malvaux L, Bovy N, Deroanne C, Lambert V, Gonzalez ML, Colige A, Rakic JM, Noel A, Martial JA, et al. MicroRNA-21 exhibits antiangiogenic function by targeting RhoB expression in endothelial cells. PloS One 2011; 6(2):e16979; PMID:21347332; https://doi.org/10.1371/journal.pone.0016979
  • Gottesbuhren U, Garg R, Riou P, McColl B, Brayson D, Ridley AJ. Rnd3 induces stress fibres in endothelial cells through RhoB. Biol Open 2013; 2(2):210-6; PMID:23430146; https://doi.org/10.1242/bio.20123574
  • Huang M, Prendergast GC. RhoB in cancer suppression. Histol Histopathol 2006; 21(2):213-8; PMID:16329046
  • Vega FM, Ridley AJ. Rho GTPases in cancer cell biology. FEBS Lett 2008; 582(14):2093-101; PMID:18460342; https://doi.org/10.1016/j.febslet.2008.04.039
  • Karlsson R, Pedersen ED, Wang Z, Brakebusch C. Rho GTPase function in tumorigenesis. Biochim Biophys Acta 2009; 1796(2):91-8; PMID:19327386
  • Ridley AJ. RhoA, RhoB and RhoC have different roles in cancer cell migration. J Microsc 2013; 251(3):242-9; PMID:23488932; https://doi.org/10.1111/jmi.12025
  • Prendergast GC, Khosravi-Far R, Solski PA, Kurzawa H, Lebowitz PF, Der CJ. Critical role of Rho in cell transformation by oncogenic Ras. Oncogene 1995; 10(12):2289-96; PMID:7784077
  • Liu A, Cerniglia GJ, Bernhard EJ, Prendergast GC. RhoB is required to mediate apoptosis in neoplastically transformed cells after DNA damage. Proc Natl Acad Sci U S A 2001; 98(11):6192-7; PMID:11353846; https://doi.org/10.1073/pnas.111137198
  • Marlow LA, Reynolds LA, Cleland AS, Cooper SJ, Gumz ML, Kurakata S, Fujiwara K, Zhang Y, Sebo T, Grant C, et al. Reactivation of suppressed RhoB is a critical step for the inhibition of anaplastic thyroid cancer growth. Cancer Res 2009; 69(4):1536-44; PMID:19208833; https://doi.org/10.1158/0008-5472.CAN-08-3718
  • Chen Z, Sun J, Pradines A, Favre G, Adnane J, Sebti SM. Both farnesylated and geranylgeranylated RhoB inhibit malignant transformation and suppress human tumor growth in nude mice. J Biol Chem 2000; 275(24):17974-8; PMID:10770919; https://doi.org/10.1074/jbc.C000145200
  • Jiang K, Sun J, Cheng J, Djeu JY, Wei S, Sebti S. Akt mediates Ras downregulation of RhoB, a suppressor of transformation, invasion, and metastasis. Mol Cell Biol 2004; 24(12):5565-76; PMID:15169915; https://doi.org/10.1128/MCB.24.12.5565-5576.2004
  • Hutchison N, Hendry BM, Sharpe CC. Rho isoforms have distinct and specific functions in the process of epithelial to mesenchymal transition in renal proximal tubular cells. Cell Signal 2009; 21(10):1522-31; PMID:19477269; https://doi.org/10.1016/j.cellsig.2009.05.012
  • Ma Y, Gong Y, Cheng Z, Loganathan S, Kao C, Sarkaria JN, Abel TW, Wang J. Critical functions of RhoB in support of glioblastoma tumorigenesis. Neuro Oncol 2015; 17(4):516-25; PMID:25216671; https://doi.org/10.1093/neuonc/nou228
  • Westmark CJ, Bartleson VB, Malter JS. RhoB mRNA is stabilized by HuR after UV light. Oncogene 2005; 24(3):502-11; PMID:15543229; https://doi.org/10.1038/sj.onc.1208224
  • Mamouni K, Cristini A, Guirouilh-Barbat J, Monferran S, Lemarie A, Faye JC, Lopez BS, Favre G, Sordet O. RhoB promotes gammaH2AX dephosphorylation and DNA double-strand break repair. Mol Cell Biol 2014; 34(16):3144-55; PMID:24912678; https://doi.org/10.1128/MCB.01525-13
  • Mazieres J, Tovar D, He B, Nieto-Acosta J, Marty-Detraves C, Clanet C, Pradines A, Jablons D, Favre G. Epigenetic regulation of RhoB loss of expression in lung cancer. BMC Cancer 2007; 7:220; PMID:18047684; https://doi.org/10.1186/1471-2407-7-220
  • Wang S, Yan-Neale Y, Fischer D, Zeremski M, Cai R, Zhu J, Asselbergs F, Hampton G, Cohen D. Histone deacetylase 1 represses the small GTPase RhoB expression in human nonsmall lung carcinoma cell line. Oncogene 2003; 22(40):6204-13; PMID:13679859; https://doi.org/10.1038/sj.onc.1206653
  • Ferreira AC, de-Freitas-Junior JC, Morgado-Diaz JA, Ridley AJ, Klumb CE. Dual inhibition of histone deacetylases and phosphoinositide 3-kinases: effects on Burkitt lymphoma cell growth and migration. J Leukoc Biol 2016; 99(4):569-578; PMID:26561567; https://doi.org/10.1189/jlb.2A0415-162R
  • Marlow LA, Bok I, Smallridge RC, Copland JA. RhoB upregulation leads to either apoptosis or cytostasis through differential target selection. Endocrine Related Cancer 2015; 22(5):777-92; PMID:26206775; https://doi.org/10.1530/ERC-14-0302
  • Jiang K, Delarue FL, Sebti SM. EGFR, ErbB2 and Ras but not Src suppress RhoB expression while ectopic expression of RhoB antagonizes oncogene-mediated transformation. Oncogene 2004; 23(5):1136-45; PMID:14647415; https://doi.org/10.1038/sj.onc.1207236
  • Ichijo S, Furuya F, Shimura H, Hayashi Y, Takahashi K, Ohta K, Kobayashi T, Kitamura K. Activation of the RhoB signaling pathway by thyroid hormone receptor β in thyroid cancer cells. PloS One 2014; 9(12):e116252; PMID:25548921; https://doi.org/10.1371/journal.pone.0116252
  • Srougi MC, Burridge K. The nuclear guanine nucleotide exchange factors Ect2 and Net1 regulate RhoB-mediated cell death after DNA damage. PloS One 2011; 6(2):e17108; PMID:21373644; https://doi.org/10.1371/journal.pone.0017108
  • Mazieres J, Tillement V, Allal C, Clanet C, Bobin L, Chen Z, Sebti SM, Favre G, Pradines A. Geranylgeranylated, but not farnesylated, RhoB suppresses Ras transformation of NIH-3T3 cells. Exp Cell Res 2005; 304(2):354-64; PMID:15748883; https://doi.org/10.1016/j.yexcr.2004.10.019
  • Kamasani U, Huang M, Duhadaway JB, Prochownik EV, Donover PS, Prendergast GC. Cyclin B1 is a critical target of RhoB in the cell suicide program triggered by farnesyl transferase inhibition. Cancer Res 2004; 64(22):8389-96; PMID:15548709; https://doi.org/10.1158/0008-5472.CAN-04-2437
  • Kamasani U, Liu AX, Prendergast GC. Genetic response to farnesyltransferase inhibitors: proapoptotic targets of RhoB. Cancer Biol Ther 2003; 2(3):273-80; PMID:12878865; https://doi.org/10.4161/cbt.2.3.385
  • Vega FM, Colomba A, Reymond N, Thomas M, Ridley AJ. RhoB regulates cell migration through altered focal adhesion dynamics. Open Biol 2012; 2(5):120076; PMID:22724071; https://doi.org/10.1098/rsob.120076
  • Alfano D, Ragno P, Stoppelli MP, Ridley AJ. RhoB regulates uPAR signalling. J Cell Sci 2012; 125(Pt 10):2369-80; PMID:22366462; https://doi.org/10.1242/jcs.091579
  • Huang M, Satchell L, Duhadaway JB, Prendergast GC, Laury-Kleintop LD. RhoB links PDGF signaling to cell migration by coordinating activation and localization of Cdc42 and Rac. J Cell Biochem 2011; 112(6):1572-84; PMID:21344485; https://doi.org/10.1002/jcb.23069
  • Vega FM, Thomas M, Reymond N, Ridley AJ. The Rho GTPase RhoB regulates cadherin expression and epithelial cell-cell interaction. Cell Commun Signal 2015; 13:6; PMID:25630770; https://doi.org/10.1186/s12964-015-0085-y
  • Bousquet E, Calvayrac O, Mazieres J, Lajoie-Mazenc I, Boubekeur N, Favre G, Pradines A. RhoB loss induces Rac1-dependent mesenchymal cell invasion in lung cells through PP2A inhibition. Oncogene 2015; 35:1760-9; PMID:26148238