The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression

Detalhes bibliográficos
Autor(a) principal: Kotelevets, Larissa
Data de Publicação: 2018
Outros Autores: Walker, Francine, Mamadou, Godefroy, Lehy, Thérèse, Jordan, Peter, Chastre, Eric
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.18/5799
Resumo: 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.
id RCAP_18d70577037cb284017f5b819eb7fb59
oai_identifier_str oai:repositorio.insa.pt:10400.18/5799
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progressionIntestinal CancerCancerRAC1bMouse ModelVias de Transdução de Sinal e Patologias AssociadasWe 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.Springer NatureRepositório Científico do Instituto Nacional de SaúdeKotelevets, LarissaWalker, FrancineMamadou, GodefroyLehy, ThérèseJordan, PeterChastre, Eric2019-02-13T17:00:06Z2018-112018-11-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.18/5799engOncogene. 2018 Nov;37(46):6054-6068. doi: 10.1038/s41388-018-0389-7. Epub 2018 Jul 9.0950-923210.1038/s41388-018-0389-7info:eu-repo/semantics/embargoedAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-07-20T15:41:04Zoai:repositorio.insa.pt:10400.18/5799Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T18:40:30.085925Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
title The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
spellingShingle The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
Kotelevets, Larissa
Intestinal Cancer
Cancer
RAC1b
Mouse Model
Vias de Transdução de Sinal e Patologias Associadas
title_short The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
title_full The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
title_fullStr The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
title_full_unstemmed The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
title_sort The Rac1 splice form Rac1b favors mouse colonic mucosa regeneration and contributes to intestinal cancer progression
author Kotelevets, Larissa
author_facet Kotelevets, Larissa
Walker, Francine
Mamadou, Godefroy
Lehy, Thérèse
Jordan, Peter
Chastre, Eric
author_role author
author2 Walker, Francine
Mamadou, Godefroy
Lehy, Thérèse
Jordan, Peter
Chastre, Eric
author2_role author
author
author
author
author
dc.contributor.none.fl_str_mv Repositório Científico do Instituto Nacional de Saúde
dc.contributor.author.fl_str_mv Kotelevets, Larissa
Walker, Francine
Mamadou, Godefroy
Lehy, Thérèse
Jordan, Peter
Chastre, Eric
dc.subject.por.fl_str_mv Intestinal Cancer
Cancer
RAC1b
Mouse Model
Vias de Transdução de Sinal e Patologias Associadas
topic Intestinal Cancer
Cancer
RAC1b
Mouse Model
Vias de Transdução de Sinal e Patologias Associadas
description 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.
publishDate 2018
dc.date.none.fl_str_mv 2018-11
2018-11-01T00:00:00Z
2019-02-13T17:00:06Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10400.18/5799
url http://hdl.handle.net/10400.18/5799
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv Oncogene. 2018 Nov;37(46):6054-6068. doi: 10.1038/s41388-018-0389-7. Epub 2018 Jul 9.
0950-9232
10.1038/s41388-018-0389-7
dc.rights.driver.fl_str_mv info:eu-repo/semantics/embargoedAccess
eu_rights_str_mv embargoedAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Springer Nature
publisher.none.fl_str_mv Springer Nature
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799132147086262272