Cargando…

Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma

Non-small cell lung cancer (NSCLC), including adenocarcinoma and squamous cell carcinoma, is the leading cause of death from lung malignancies and has a poor prognosis due to metastasis. Suppressor of cytokine signalling-2 (SOCS2), a feedback inhibitor of cytokine signalling, has been shown to be in...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Yue, Zhang, Zhilei, Wang, Ning, Chen, Jizheng, Zhang, Xu, Guo, Min, John Zhong, Li, Wang, Qian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5861121/
https://www.ncbi.nlm.nih.gov/pubmed/29559623
http://dx.doi.org/10.1038/s41419-018-0457-5
_version_ 1783308037100929024
author Zhou, Yue
Zhang, Zhilei
Wang, Ning
Chen, Jizheng
Zhang, Xu
Guo, Min
John Zhong, Li
Wang, Qian
author_facet Zhou, Yue
Zhang, Zhilei
Wang, Ning
Chen, Jizheng
Zhang, Xu
Guo, Min
John Zhong, Li
Wang, Qian
author_sort Zhou, Yue
collection PubMed
description Non-small cell lung cancer (NSCLC), including adenocarcinoma and squamous cell carcinoma, is the leading cause of death from lung malignancies and has a poor prognosis due to metastasis. Suppressor of cytokine signalling-2 (SOCS2), a feedback inhibitor of cytokine signalling, has been shown to be involved in growth control. Here, we show that SOCS2 were significantly downregulated in tumour foci in NSCLC patients. The expression levels of SOCS2 significantly correlated with clinical stage, lymph node metastasis, histological subtype and survival time. In particular, the decreased expression of SOCS2 significantly associated with advanced pathological stage, lymph node metastasis and shorter overall survival in lung adenocarcinoma patients. In vivo animal results showed that overexpressed SOCS2 attenuated the metastatic characteristics of lung adenocarcinoma, including by inhibiting the epithelial–mesenchymal transition (EMT). Further functional studies indicated that insulin-like growth factor 1 (IGF1)-driven migratory and invasive behaviours of lung adenocarcinoma cells can be partially suppressed by exogenous SOCS2 expression. Investigations into the mechanism of action revealed that SOCS2 inhibits EMT by inactivating signal transducer and activator of transcription 3 (STAT3) and STAT5 via the competitive binding of SOCS2 to the STAT binding sites on IGF1R. Altogether, our results reveal an important role for SOCS2 dysregulation in the pathogenicity of lung adenocarcinoma, suggest its potential use as a biomarker for diagnosing lung adenocarcinoma, and paves the way to develop novel therapy targets as the axis of SOCS2–IGF1R–STAT in lung adenocarcinoma.
format Online
Article
Text
id pubmed-5861121
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-58611212018-06-04 Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma Zhou, Yue Zhang, Zhilei Wang, Ning Chen, Jizheng Zhang, Xu Guo, Min John Zhong, Li Wang, Qian Cell Death Dis Article Non-small cell lung cancer (NSCLC), including adenocarcinoma and squamous cell carcinoma, is the leading cause of death from lung malignancies and has a poor prognosis due to metastasis. Suppressor of cytokine signalling-2 (SOCS2), a feedback inhibitor of cytokine signalling, has been shown to be involved in growth control. Here, we show that SOCS2 were significantly downregulated in tumour foci in NSCLC patients. The expression levels of SOCS2 significantly correlated with clinical stage, lymph node metastasis, histological subtype and survival time. In particular, the decreased expression of SOCS2 significantly associated with advanced pathological stage, lymph node metastasis and shorter overall survival in lung adenocarcinoma patients. In vivo animal results showed that overexpressed SOCS2 attenuated the metastatic characteristics of lung adenocarcinoma, including by inhibiting the epithelial–mesenchymal transition (EMT). Further functional studies indicated that insulin-like growth factor 1 (IGF1)-driven migratory and invasive behaviours of lung adenocarcinoma cells can be partially suppressed by exogenous SOCS2 expression. Investigations into the mechanism of action revealed that SOCS2 inhibits EMT by inactivating signal transducer and activator of transcription 3 (STAT3) and STAT5 via the competitive binding of SOCS2 to the STAT binding sites on IGF1R. Altogether, our results reveal an important role for SOCS2 dysregulation in the pathogenicity of lung adenocarcinoma, suggest its potential use as a biomarker for diagnosing lung adenocarcinoma, and paves the way to develop novel therapy targets as the axis of SOCS2–IGF1R–STAT in lung adenocarcinoma. Nature Publishing Group UK 2018-03-20 /pmc/articles/PMC5861121/ /pubmed/29559623 http://dx.doi.org/10.1038/s41419-018-0457-5 Text en © The Author(s) 2018 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/.
spellingShingle Article
Zhou, Yue
Zhang, Zhilei
Wang, Ning
Chen, Jizheng
Zhang, Xu
Guo, Min
John Zhong, Li
Wang, Qian
Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title_full Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title_fullStr Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title_full_unstemmed Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title_short Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
title_sort suppressor of cytokine signalling-2 limits igf1r-mediated regulation of epithelial–mesenchymal transition in lung adenocarcinoma
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5861121/
https://www.ncbi.nlm.nih.gov/pubmed/29559623
http://dx.doi.org/10.1038/s41419-018-0457-5
work_keys_str_mv AT zhouyue suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT zhangzhilei suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT wangning suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT chenjizheng suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT zhangxu suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT guomin suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT johnzhongli suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma
AT wangqian suppressorofcytokinesignalling2limitsigf1rmediatedregulationofepithelialmesenchymaltransitioninlungadenocarcinoma