RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells

Detalhes bibliográficos
Autor(a) principal: Faria, Márcia
Data de Publicação: 2017
Outros Autores: Matos, Paulo, Pereira, Teresa, Cabrera, Rafael, Cardoso, Bruno A., Bugalho, Maria João, Silva, Ana Luísa
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.18/5458
Resumo: Overexpression of tumor-associated RAC1b has been recently highlighted as one of the most promising targets for therapeutic intervention in colon, breast, lung and pancreatic cancer. RAC1b is a hyperactive variant of the small GTPase RAC1 and has been recently shown to be overexpressed in a subset of papillary thyroid carcinomas associated with unfavorable outcome. Using the K1 PTC derived cell line as an in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-kB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-kB canonical pathway activation. Consistently, we observed a RAC1b-mediated decrease in IκBα (NF-kB inhibitor) protein levels. Moreover, we show that RAC1b overexpression stimulates G1/S progression and protects thyroid cells against induced apoptosis, the latter through a process involving the NF-kB pathway. Present data support previous findings suggesting an important role for RAC1b in the development of follicular cell-derived thyroid malignancies and point out NF-kB activation as one of the molecular mechanisms associated with the pro-tumorigenic advantage of RAC1b overexpression in thyroid carcinomas.
id RCAP_8477a33971c078288c3b8073e6e3b54d
oai_identifier_str oai:repositorio.insa.pt:10400.18/5458
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cellsApoptosisCell Line, TumorCell ProliferationCyclin D1G1 Phase Cell Cycle CheckpointsHumansNF-KappaB Inhibitor alphaNF-kappa BSignal TransductionStaurosporineThyroid Epithelial CellsTransfectionrac1 GTP-Binding ProteinGene Expression Regulation, NeoplasticVias de Transdução de Sinal e Patologias AssociadasOverexpression of tumor-associated RAC1b has been recently highlighted as one of the most promising targets for therapeutic intervention in colon, breast, lung and pancreatic cancer. RAC1b is a hyperactive variant of the small GTPase RAC1 and has been recently shown to be overexpressed in a subset of papillary thyroid carcinomas associated with unfavorable outcome. Using the K1 PTC derived cell line as an in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-kB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-kB canonical pathway activation. Consistently, we observed a RAC1b-mediated decrease in IκBα (NF-kB inhibitor) protein levels. Moreover, we show that RAC1b overexpression stimulates G1/S progression and protects thyroid cells against induced apoptosis, the latter through a process involving the NF-kB pathway. Present data support previous findings suggesting an important role for RAC1b in the development of follicular cell-derived thyroid malignancies and point out NF-kB activation as one of the molecular mechanisms associated with the pro-tumorigenic advantage of RAC1b overexpression in thyroid carcinomas.Public Library of ScienceRepositório Científico do Instituto Nacional de SaúdeFaria, MárciaMatos, PauloPereira, TeresaCabrera, RafaelCardoso, Bruno A.Bugalho, Maria JoãoSilva, Ana Luísa2018-03-22T19:09:09Z2017-02-242017-02-24T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.18/5458engPLoS One. 2017 Feb 24;12(2):e0172689. doi: 10.1371/journal.pone.0172689. eCollection 2017.1932-6203)10.1371/journal.pone.0172689info:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-07-20T15:40:52Zoai:repositorio.insa.pt:10400.18/5458Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T18:40:08.944876Repositório 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 RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
title RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
spellingShingle RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
Faria, Márcia
Apoptosis
Cell Line, Tumor
Cell Proliferation
Cyclin D1
G1 Phase Cell Cycle Checkpoints
Humans
NF-KappaB Inhibitor alpha
NF-kappa B
Signal Transduction
Staurosporine
Thyroid Epithelial Cells
Transfection
rac1 GTP-Binding Protein
Gene Expression Regulation, Neoplastic
Vias de Transdução de Sinal e Patologias Associadas
title_short RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
title_full RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
title_fullStr RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
title_full_unstemmed RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
title_sort RAC1b overexpression stimulates proliferation and NF-kB-mediated anti-apoptotic signaling in thyroid cancer cells
author Faria, Márcia
author_facet Faria, Márcia
Matos, Paulo
Pereira, Teresa
Cabrera, Rafael
Cardoso, Bruno A.
Bugalho, Maria João
Silva, Ana Luísa
author_role author
author2 Matos, Paulo
Pereira, Teresa
Cabrera, Rafael
Cardoso, Bruno A.
Bugalho, Maria João
Silva, Ana Luísa
author2_role author
author
author
author
author
author
dc.contributor.none.fl_str_mv Repositório Científico do Instituto Nacional de Saúde
dc.contributor.author.fl_str_mv Faria, Márcia
Matos, Paulo
Pereira, Teresa
Cabrera, Rafael
Cardoso, Bruno A.
Bugalho, Maria João
Silva, Ana Luísa
dc.subject.por.fl_str_mv Apoptosis
Cell Line, Tumor
Cell Proliferation
Cyclin D1
G1 Phase Cell Cycle Checkpoints
Humans
NF-KappaB Inhibitor alpha
NF-kappa B
Signal Transduction
Staurosporine
Thyroid Epithelial Cells
Transfection
rac1 GTP-Binding Protein
Gene Expression Regulation, Neoplastic
Vias de Transdução de Sinal e Patologias Associadas
topic Apoptosis
Cell Line, Tumor
Cell Proliferation
Cyclin D1
G1 Phase Cell Cycle Checkpoints
Humans
NF-KappaB Inhibitor alpha
NF-kappa B
Signal Transduction
Staurosporine
Thyroid Epithelial Cells
Transfection
rac1 GTP-Binding Protein
Gene Expression Regulation, Neoplastic
Vias de Transdução de Sinal e Patologias Associadas
description Overexpression of tumor-associated RAC1b has been recently highlighted as one of the most promising targets for therapeutic intervention in colon, breast, lung and pancreatic cancer. RAC1b is a hyperactive variant of the small GTPase RAC1 and has been recently shown to be overexpressed in a subset of papillary thyroid carcinomas associated with unfavorable outcome. Using the K1 PTC derived cell line as an in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-kB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-kB canonical pathway activation. Consistently, we observed a RAC1b-mediated decrease in IκBα (NF-kB inhibitor) protein levels. Moreover, we show that RAC1b overexpression stimulates G1/S progression and protects thyroid cells against induced apoptosis, the latter through a process involving the NF-kB pathway. Present data support previous findings suggesting an important role for RAC1b in the development of follicular cell-derived thyroid malignancies and point out NF-kB activation as one of the molecular mechanisms associated with the pro-tumorigenic advantage of RAC1b overexpression in thyroid carcinomas.
publishDate 2017
dc.date.none.fl_str_mv 2017-02-24
2017-02-24T00:00:00Z
2018-03-22T19:09:09Z
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/10400.18/5458
url http://hdl.handle.net/10400.18/5458
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv PLoS One. 2017 Feb 24;12(2):e0172689. doi: 10.1371/journal.pone.0172689. eCollection 2017.
1932-6203)
10.1371/journal.pone.0172689
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Public Library of Science
publisher.none.fl_str_mv Public Library of Science
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_ 1799132143232745472