000 02680na a2200229 4500
003 PC4214
005 20210625062801.0
008 130622s2011 xxx||||| |||| 00| 0 eng d
040 _cH12O
041 _aeng
100 _aMedraño Fernández, Iria
_91623
_eInstituto de Investigación i+12
245 0 0 _aRap1-GTP-interacting Adaptor Molecule (RIAM) protein controls invasion and growth of melanoma cells
_h[artículo]
260 _bJournal of Biological Chemistry,
_c2011
300 _a286(21):18492-18504.
500 _aFormato Vancouver: Hernández-Varas P, Coló GP, Bartolomé RA, Paterson A, Medraño-Fernández I, Arellano-Sánchez N, et al. Rap1-GTP-interacting adaptor molecule (RIAM) protein controls invasion and growth of melanoma cells. J Biol Chem. 2011;286(21):18492-504.
501 _aPMID: 21454517
504 _aContiene 38 referencias.
520 _aThe Mig-10/RIAM/lamellipodin (MRL) family member Rap1-GTP-interacting adaptor molecule (RIAM) interacts with active Rap1, a small GTPase that is frequently activated in tumors such as melanoma and prostate cancer. We show here that RIAM is expressed in metastatic human melanoma cells and that both RIAM and Rap1 are required for BLM melanoma cell invasion. RIAM silencing in melanoma cells led to inhibition of tumor growth and to delayed metastasis in a severe combined immunodeficiency xenograft model. Defective invasion of RIAM-silenced melanoma cells arose from impairment in persistent cell migration directionality, which was associated with deficient activation of a Vav2-RhoA-ROCK-myosin light chain pathway. Expression of constitutively active Vav2 and RhoA in cells depleted for RIAM partially rescued their invasion, indicating that Vav2 and RhoA mediate RIAM function. These results suggest that inhibition of cell invasion in RIAM-silenced melanoma cells is likely based on altered cell contractility and cell polarization. Furthermore, we show that RIAM depletion reduces beta 1 integrin-dependent melanoma cell adhesion, which correlates with decreased activation of both Erk1/2 MAPK and phosphatidylinositol 3-kinase, two central molecules controlling cell growth and cell survival. In addition to causing inhibition of cell proliferation, RIAM silencing led to higher susceptibility to cell apoptosis. Together, these data suggest that defective activation of these kinases in RIAM-silenced cells could account for inhibition of melanoma cell growth and that RIAM might contribute to the dissemination of melanoma cells.
710 _9625
_aInstituto de Investigación imas12
856 _uhttp://pc-h12o-es.m-hdoct.a17.csinet.es/pdf/pc/4/pc4214.pdf
_ySolicitar documento
942 _2ddc
_cART
_n0
999 _c4214
_d4214