Your browser doesn't support javascript.
loading
Autocrine netrin function inhibits glioma cell motility and promotes focal adhesion formation.
Jarjour, Andrew A; Durko, Margaret; Luk, Tamarah L; Marçal, Nathalie; Shekarabi, Masoud; Kennedy, Timothy E.
Afiliação
  • Jarjour AA; Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
PLoS One ; 6(9): e25408, 2011.
Article em En | MEDLINE | ID: mdl-21980448
ABSTRACT
Deregulation of mechanisms that control cell motility plays a key role in tumor progression by promoting tumor cell dissemination. Secreted netrins and their receptors, Deleted in Colorectal Cancer (DCC), neogenin, and the UNC5 homologues, regulate cell and axon migration, cell adhesion, and tissue morphogenesis. Netrin and netrin receptor expression have previously been shown to be disrupted in invasive tumors, including glioblastoma. We determined that the human glioblastoma cell lines U87, U343, and U373 all express neogenin, UNC5 homologues, and netrin-1 or netrin-3, but only U87 cells express DCC. Using transfilter migration assays, we demonstrate DCC-dependent chemoattractant migration of U87 cells up a gradient of netrin-1. In contrast, U343 and U373 cells, which do not express DCC, were neither attracted nor repelled. Ectopic expression of DCC by U343 and U373 cells resulted in these cells becoming competent to respond to a gradient of netrin-1 as a chemoattractant, and also slowed their rate of spontaneous migration. Here, in addition to netrins' well-characterized chemotropic activity, we demonstrate an autocrine function for netrin-1 and netrin-3 in U87 and U373 cells that slows migration. We provide evidence that netrins promote the maturation of focal complexes, structures associated with cell movement, into focal adhesions. Consistent with this, netrin, DCC, and UNC5 homologues were associated with focal adhesions, but not focal complexes. Disrupting netrin or DCC function did not alter cell proliferation or survival. Our findings provide evidence that DCC can slow cell migration, and that neogenin and UNC5 homologues are not sufficient to substitute for DCC function in these cells. Furthermore, we identify a role for netrins as autocrine inhibitors of cell motility that promote focal adhesion formation. These findings suggest that disruption of netrin signalling may disable a mechanism that normally restrains inappropriate cell migration.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Movimento Celular / Comunicação Autócrina / Adesões Focais / Proteínas Supressoras de Tumor / Glioma / Fatores de Crescimento Neural Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Ano de publicação: 2011 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Movimento Celular / Comunicação Autócrina / Adesões Focais / Proteínas Supressoras de Tumor / Glioma / Fatores de Crescimento Neural Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Ano de publicação: 2011 Tipo de documento: Article