Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?

Detalhes bibliográficos
Autor(a) principal: Faria, Márcia Carina da Silva
Data de Publicação: 2015
Tipo de documento: Dissertação
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10362/61263
Resumo: Papillary and Follicular thyroid carcinomas (PTC and FTC) are lesions derived from follicular cells. The standard therapy includes surgery and radioiodine treatment. Despite being usually associated with a good prognosis the rate of recurrence is high and a subset of patients present radioiodine refractory and non-responsive neoplasia. So, the search for new therapeutic alternatives, as well as new diagnostic/prognostic markers is highly relevant. Our group at IPOLFG has previously shown RAC1b to be overexpressed in PTCs, being this overexpression significantly associated with poorer outcomes. RAC1b is a variant of the small GTPase RAC1 that was shown to have the ability to stimulate the canonical NF-κB regulatory pathway in colorectal cancer. NF-κB activation is associated with the tumorigenic process in several types of cancer, mainly due to its role in apoptosis evasion. Here, we aimed to extend the study of the role of RAC1b to FTCs and further investigate the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis. Using the human papillary thyroid carcinoma derived cell line as in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-κB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-κB canonical pathway activation. Consistently, we were able to observe a RAC1b-mediated decrease in IκBα (NF-κB inhibitor) protein levels. These findings prompted us to further assess the cellular consequences of RAC1b-mediated NF-κB activation in a thyroid biological system. Our results indicated that RAC1b protected cells against apoptosis and stimulated G1/S progression, through a process involving the NF-κB pathway. Summing up, presented findings suggest an important role of RAC1b in the progression of follicular cell derived thyroid carcinomas and point out NF-κB activation as one of the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis.
id RCAP_25006366129935892d0ed931ac9e73fb
oai_identifier_str oai:run.unl.pt:10362/61263
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?Thyroid tumorigenesisRAC1bNF-κB canonical pathwayCyclin D1ApoptosisCell CycleDomínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e TecnologiasPapillary and Follicular thyroid carcinomas (PTC and FTC) are lesions derived from follicular cells. The standard therapy includes surgery and radioiodine treatment. Despite being usually associated with a good prognosis the rate of recurrence is high and a subset of patients present radioiodine refractory and non-responsive neoplasia. So, the search for new therapeutic alternatives, as well as new diagnostic/prognostic markers is highly relevant. Our group at IPOLFG has previously shown RAC1b to be overexpressed in PTCs, being this overexpression significantly associated with poorer outcomes. RAC1b is a variant of the small GTPase RAC1 that was shown to have the ability to stimulate the canonical NF-κB regulatory pathway in colorectal cancer. NF-κB activation is associated with the tumorigenic process in several types of cancer, mainly due to its role in apoptosis evasion. Here, we aimed to extend the study of the role of RAC1b to FTCs and further investigate the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis. Using the human papillary thyroid carcinoma derived cell line as in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-κB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-κB canonical pathway activation. Consistently, we were able to observe a RAC1b-mediated decrease in IκBα (NF-κB inhibitor) protein levels. These findings prompted us to further assess the cellular consequences of RAC1b-mediated NF-κB activation in a thyroid biological system. Our results indicated that RAC1b protected cells against apoptosis and stimulated G1/S progression, through a process involving the NF-κB pathway. Summing up, presented findings suggest an important role of RAC1b in the progression of follicular cell derived thyroid carcinomas and point out NF-κB activation as one of the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis.Silva, AnaRUNFaria, Márcia Carina da Silva2019-02-22T11:24:06Z2015-0920152015-09-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/masterThesisapplication/pdfhttp://hdl.handle.net/10362/61263enginfo: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:RCAAP2024-03-11T04:29:08Zoai:run.unl.pt:10362/61263Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-20T03:33:36.251533Repositório 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 Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
title Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
spellingShingle Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
Faria, Márcia Carina da Silva
Thyroid tumorigenesis
RAC1b
NF-κB canonical pathway
Cyclin D1
Apoptosis
Cell Cycle
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
title_short Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
title_full Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
title_fullStr Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
title_full_unstemmed Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
title_sort Overexpression of RAC1b in follicular cell derived thyroid cancers: An activator of NF-κB pathway that contributes to thyroid tumorigenesis?
author Faria, Márcia Carina da Silva
author_facet Faria, Márcia Carina da Silva
author_role author
dc.contributor.none.fl_str_mv Silva, Ana
RUN
dc.contributor.author.fl_str_mv Faria, Márcia Carina da Silva
dc.subject.por.fl_str_mv Thyroid tumorigenesis
RAC1b
NF-κB canonical pathway
Cyclin D1
Apoptosis
Cell Cycle
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
topic Thyroid tumorigenesis
RAC1b
NF-κB canonical pathway
Cyclin D1
Apoptosis
Cell Cycle
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
description Papillary and Follicular thyroid carcinomas (PTC and FTC) are lesions derived from follicular cells. The standard therapy includes surgery and radioiodine treatment. Despite being usually associated with a good prognosis the rate of recurrence is high and a subset of patients present radioiodine refractory and non-responsive neoplasia. So, the search for new therapeutic alternatives, as well as new diagnostic/prognostic markers is highly relevant. Our group at IPOLFG has previously shown RAC1b to be overexpressed in PTCs, being this overexpression significantly associated with poorer outcomes. RAC1b is a variant of the small GTPase RAC1 that was shown to have the ability to stimulate the canonical NF-κB regulatory pathway in colorectal cancer. NF-κB activation is associated with the tumorigenic process in several types of cancer, mainly due to its role in apoptosis evasion. Here, we aimed to extend the study of the role of RAC1b to FTCs and further investigate the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis. Using the human papillary thyroid carcinoma derived cell line as in vitro model, we observed that both RAC1 and RAC1b were able to induce a significant increase on NF-κB and cyclin D1 reporter activity. A clear p65 nuclear localization was found in cells transfected with RAC1b-WT, confirming NF-κB canonical pathway activation. Consistently, we were able to observe a RAC1b-mediated decrease in IκBα (NF-κB inhibitor) protein levels. These findings prompted us to further assess the cellular consequences of RAC1b-mediated NF-κB activation in a thyroid biological system. Our results indicated that RAC1b protected cells against apoptosis and stimulated G1/S progression, through a process involving the NF-κB pathway. Summing up, presented findings suggest an important role of RAC1b in the progression of follicular cell derived thyroid carcinomas and point out NF-κB activation as one of the molecular mechanisms associated with RAC1b overexpression and downstream signalling in thyroid tumorigenesis.
publishDate 2015
dc.date.none.fl_str_mv 2015-09
2015
2015-09-01T00:00:00Z
2019-02-22T11:24:06Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/masterThesis
format masterThesis
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10362/61263
url http://hdl.handle.net/10362/61263
dc.language.iso.fl_str_mv eng
language eng
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.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_ 1799137957645385728