Your browser doesn't support javascript.
loading
The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression.
Kotelevets, Larissa; Walker, Francine; Mamadou, Godefroy; Lehy, Thérèse; Jordan, Peter; Chastre, Eric.
Afiliação
  • Kotelevets L; Institut National de la Santé et de la Recherche Médicale, UMR S 938, Centre de Recherche Saint-Antoine, Paris, France. larissa.kotelevets@inserm.fr.
  • Walker F; Université Pierre-et-Marie-Curie Paris 6, Hôpital Saint-Antoine, Site Bâtiment Kourilsky, Paris, France. larissa.kotelevets@inserm.fr.
  • Mamadou G; Institut National de la Santé et de la Recherche Médicale, U1149, Centre National de la Recherche Scientifique-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.
  • Lehy T; Assistance publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val de Seine Bichat-Claude Bernard, Department of Pathology, Paris, France.
  • Jordan P; TransCell-Lab Laboratory, Faculty of Medicine Xavier Bichat, University of Paris Diderot - Paris7, Paris, France.
  • Chastre E; Institut National de la Santé et de la Recherche Médicale, U1149, Centre National de la Recherche Scientifique-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.
Oncogene ; 37(46): 6054-6068, 2018 11.
Article em En | MEDLINE | ID: mdl-29985482
ABSTRACT
We previously have identified the ectopic expression of Rac1b, an activated and novel splice variant of Rac1, in a subset of human colorectal adenocarcinomas, as well as in inflammatory bowel diseases and in colitis mouse model. Rac1b overexpression has been further evidenced in breast, pancreatic, thyroid, ovarian, and lung cancers. In this context, the aim of our study was to investigate the physiopathological implications of Rac1b in intestinal inflammation and carcinogenesis in vivo. The ectopic expression of Rac1b was induced in mouse intestinal epithelial cells after crossing Rosa26-LSL-Rac1b and villin-Cre mice. These animals were let to age or were challenged with dextran sulfate sodium (DSS) to induce experimental colitis, or either received azoxymethane (AOM)/DSS treatment, or were bred with ApcMin/+ or Il10-/- mice to trigger intestinal tumors. Rac1b ectopic expression increased the intestinal epithelial cell proliferation and migration, enhanced the production of reactive oxygen species, and promoted the Paneth cell lineage. Although Rac1b overexpression alone was not sufficient to drive intestinal neoplasia, it enhanced Apc-dependent intestinal tumorigenesis. In the context of Il10 knockout, the Rac1b transgene strengthened colonic inflammation due to induced intestinal mucosa permeability and promoted cecum and proximal colon carcinogenesis. In contrast, Rac1b alleviated carcinogen/acute inflammation-associated colon carcinogenesis (AOM/DSS). This resulted at least partly from the early mucosal repair after resolution of inflammation. Our data highlight the critical role of Rac1b in driving wound-healing after resolution of intestinal inflammation, and in cooperating with Wnt pathway dysregulation and chronic inflammation to promote intestinal carcinogenesis.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neuropeptídeos / Colo / Neoplasias do Colo / Proteínas rac1 de Ligação ao GTP / Mucosa Intestinal Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neuropeptídeos / Colo / Neoplasias do Colo / Proteínas rac1 de Ligação ao GTP / Mucosa Intestinal Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article