Cargando…

RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4

Regulator of G-protein signaling 6 (RGS6) is a newly discovered tumor suppressor that has been shown to be protective in development of various cancers such as breast cancer and bladder cancer. But the mechanisms underlying these tumor-suppressing functions of RGS6 are not fully understood. Here, we...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Zhao, Chen, Jun, Wang, Shengjie, Sun, Zelong, Lei, Zhe, Zhang, Hong-Tao, Huang, Jie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9334288/
https://www.ncbi.nlm.nih.gov/pubmed/35902557
http://dx.doi.org/10.1038/s41419-022-05093-0
_version_ 1784759070906908672
author Wang, Zhao
Chen, Jun
Wang, Shengjie
Sun, Zelong
Lei, Zhe
Zhang, Hong-Tao
Huang, Jie
author_facet Wang, Zhao
Chen, Jun
Wang, Shengjie
Sun, Zelong
Lei, Zhe
Zhang, Hong-Tao
Huang, Jie
author_sort Wang, Zhao
collection PubMed
description Regulator of G-protein signaling 6 (RGS6) is a newly discovered tumor suppressor that has been shown to be protective in development of various cancers such as breast cancer and bladder cancer. But the mechanisms underlying these tumor-suppressing functions of RGS6 are not fully understood. Here, we discover a novel function of RGS6 in suppressing TGF-β-induced epithelial–mesenchymal transition (EMT) of non-small cell lung cancer (NSCLC) cells and in vivo NSCLC metastasis. Using both bioinformatics and experimental tools, we showed that RGS6 was downregulated in lung cancer tissues compared to noncancerous counterparts, and low expression of RGS6 was associated with poor survival of lung cancer patients. Overexpression of RGS6 suppressed TGF-β-induced EMT in vitro and TGF-β-promoted metastasis in vivo, by impairing gene expression of downstream effectors induced by the canonical TGF-β-SMAD signaling. The ability of RGS6 to suppress TGF-β-SMAD-mediated gene expression relied on its binding to SMAD4 to prevent complex formation between SMAD4 and SMAD2/3, but independent of its regulation of the G-protein signaling. Interaction between RGS6 and SMAD4 caused less nuclear entry of p-SMAD3 and SMAD4, resulting in inefficient SMAD3-mediated gene expression. Taken together, our findings reveal a novel and noncanonical role of RGS6 in regulation of TGF-β-induced EMT and metastasis of NSCLC and identify RGS6 as a prognostic marker and a potential novel target for NSCLC therapy.
format Online
Article
Text
id pubmed-9334288
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-93342882022-07-30 RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4 Wang, Zhao Chen, Jun Wang, Shengjie Sun, Zelong Lei, Zhe Zhang, Hong-Tao Huang, Jie Cell Death Dis Article Regulator of G-protein signaling 6 (RGS6) is a newly discovered tumor suppressor that has been shown to be protective in development of various cancers such as breast cancer and bladder cancer. But the mechanisms underlying these tumor-suppressing functions of RGS6 are not fully understood. Here, we discover a novel function of RGS6 in suppressing TGF-β-induced epithelial–mesenchymal transition (EMT) of non-small cell lung cancer (NSCLC) cells and in vivo NSCLC metastasis. Using both bioinformatics and experimental tools, we showed that RGS6 was downregulated in lung cancer tissues compared to noncancerous counterparts, and low expression of RGS6 was associated with poor survival of lung cancer patients. Overexpression of RGS6 suppressed TGF-β-induced EMT in vitro and TGF-β-promoted metastasis in vivo, by impairing gene expression of downstream effectors induced by the canonical TGF-β-SMAD signaling. The ability of RGS6 to suppress TGF-β-SMAD-mediated gene expression relied on its binding to SMAD4 to prevent complex formation between SMAD4 and SMAD2/3, but independent of its regulation of the G-protein signaling. Interaction between RGS6 and SMAD4 caused less nuclear entry of p-SMAD3 and SMAD4, resulting in inefficient SMAD3-mediated gene expression. Taken together, our findings reveal a novel and noncanonical role of RGS6 in regulation of TGF-β-induced EMT and metastasis of NSCLC and identify RGS6 as a prognostic marker and a potential novel target for NSCLC therapy. Nature Publishing Group UK 2022-07-28 /pmc/articles/PMC9334288/ /pubmed/35902557 http://dx.doi.org/10.1038/s41419-022-05093-0 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Wang, Zhao
Chen, Jun
Wang, Shengjie
Sun, Zelong
Lei, Zhe
Zhang, Hong-Tao
Huang, Jie
RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title_full RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title_fullStr RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title_full_unstemmed RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title_short RGS6 suppresses TGF-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4
title_sort rgs6 suppresses tgf-β-induced epithelial–mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with smad4
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9334288/
https://www.ncbi.nlm.nih.gov/pubmed/35902557
http://dx.doi.org/10.1038/s41419-022-05093-0
work_keys_str_mv AT wangzhao rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT chenjun rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT wangshengjie rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT sunzelong rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT leizhe rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT zhanghongtao rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4
AT huangjie rgs6suppressestgfbinducedepithelialmesenchymaltransitioninnonsmallcelllungcancersviaanovelmechanismdependentonitsinteractionwithsmad4