Cargando…

MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis

Background: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has a poor prognosis due to the high incidence of invasion and metastasis-related progression. However, the underlying mechanism remains elusive, and valuable biomarkers for predicting invasion, metastasis, an...

Descripción completa

Detalles Bibliográficos
Autores principales: Dian, Mei-Juan, Li, Jing, Zhang, Xiao-Ling, Li, Zi-Jian, Zhou, Ying, Zhou, Wei, Zhong, Qiu-Ling, Pang, Wen-Qian, Lin, Xiao-Lin, Liu, Tao, Liu, Yi-An, Li, Yong-Long, Han, Liu-Xin, Zhao, Wen-Tao, Jia, Jun-Shuang, Xiao, Sheng-Jun, Xiao, Dong, Xia, Jia-Wei, Hao, Wei-Chao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8210547/
https://www.ncbi.nlm.nih.gov/pubmed/34149910
http://dx.doi.org/10.7150/jca.60008
_version_ 1783709333335310336
author Dian, Mei-Juan
Li, Jing
Zhang, Xiao-Ling
Li, Zi-Jian
Zhou, Ying
Zhou, Wei
Zhong, Qiu-Ling
Pang, Wen-Qian
Lin, Xiao-Lin
Liu, Tao
Liu, Yi-An
Li, Yong-Long
Han, Liu-Xin
Zhao, Wen-Tao
Jia, Jun-Shuang
Xiao, Sheng-Jun
Xiao, Dong
Xia, Jia-Wei
Hao, Wei-Chao
author_facet Dian, Mei-Juan
Li, Jing
Zhang, Xiao-Ling
Li, Zi-Jian
Zhou, Ying
Zhou, Wei
Zhong, Qiu-Ling
Pang, Wen-Qian
Lin, Xiao-Lin
Liu, Tao
Liu, Yi-An
Li, Yong-Long
Han, Liu-Xin
Zhao, Wen-Tao
Jia, Jun-Shuang
Xiao, Sheng-Jun
Xiao, Dong
Xia, Jia-Wei
Hao, Wei-Chao
author_sort Dian, Mei-Juan
collection PubMed
description Background: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has a poor prognosis due to the high incidence of invasion and metastasis-related progression. However, the underlying mechanism remains elusive, and valuable biomarkers for predicting invasion, metastasis, and poor prognosis of HCC patients are still lacking. Methods: Immunohistochemistry (IHC) was performed on HCC tissues (n = 325), and the correlations between MST4 expression of the clinical HCC tissues, the clinicopathologic features, and survival were further evaluated. The effects of MST4 on HCC cell migratory and invasive properties in vitro were evaluated by Transwell and Boyden assays. The intrahepatic metastasis mouse model was established to evaluate the HCC metastasis in vivo. The PI3K inhibitor, LY294002, and a specific siRNA against Snail1 were used to investigate the roles of PI3K/AKT pathway and Snail1 in MST4-regulated EMT, migration, and invasion of HCC cells, respectively. Results: In this study, by comprehensively analyzing our clinical data, we discovered that low MST4 expression is highly associated with the advanced progression of HCC and serves as a prognostic biomarker for HCC patients of clinical-stage III-IV. Functional studies indicate that MST4 inactivation induces epithelial-to-mesenchymal transition (EMT) of HCC cells, promotes their migratory and invasive potential in vitro, and facilitates their intrahepatic metastasis in vivo, whereas MST4 overexpression exhibits the opposite phenotypes. Mechanistically, MST4 inactivation elevates the expression and nuclear translocation of Snail1, a key EMT transcription factor (EMT-TF), through the PI3K/AKT signaling pathway, thus inducing the EMT phenotype of HCC cells, and enhancing their invasive and metastatic potential. Moreover, a negative correlation between MST4 and p-AKT, Snail1, and Ki67 and a positive correlation between MST4 and E-cadherin were determined in clinical HCC samples. Conclusions: Our findings indicate that MST4 suppresses EMT, invasion, and metastasis of HCC cells by modulating the PI3K/AKT/Snail1 axis, suggesting that MST4 may be a potential prognostic biomarker for aggressive and metastatic HCC.
format Online
Article
Text
id pubmed-8210547
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-82105472021-06-17 MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis Dian, Mei-Juan Li, Jing Zhang, Xiao-Ling Li, Zi-Jian Zhou, Ying Zhou, Wei Zhong, Qiu-Ling Pang, Wen-Qian Lin, Xiao-Lin Liu, Tao Liu, Yi-An Li, Yong-Long Han, Liu-Xin Zhao, Wen-Tao Jia, Jun-Shuang Xiao, Sheng-Jun Xiao, Dong Xia, Jia-Wei Hao, Wei-Chao J Cancer Research Paper Background: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and has a poor prognosis due to the high incidence of invasion and metastasis-related progression. However, the underlying mechanism remains elusive, and valuable biomarkers for predicting invasion, metastasis, and poor prognosis of HCC patients are still lacking. Methods: Immunohistochemistry (IHC) was performed on HCC tissues (n = 325), and the correlations between MST4 expression of the clinical HCC tissues, the clinicopathologic features, and survival were further evaluated. The effects of MST4 on HCC cell migratory and invasive properties in vitro were evaluated by Transwell and Boyden assays. The intrahepatic metastasis mouse model was established to evaluate the HCC metastasis in vivo. The PI3K inhibitor, LY294002, and a specific siRNA against Snail1 were used to investigate the roles of PI3K/AKT pathway and Snail1 in MST4-regulated EMT, migration, and invasion of HCC cells, respectively. Results: In this study, by comprehensively analyzing our clinical data, we discovered that low MST4 expression is highly associated with the advanced progression of HCC and serves as a prognostic biomarker for HCC patients of clinical-stage III-IV. Functional studies indicate that MST4 inactivation induces epithelial-to-mesenchymal transition (EMT) of HCC cells, promotes their migratory and invasive potential in vitro, and facilitates their intrahepatic metastasis in vivo, whereas MST4 overexpression exhibits the opposite phenotypes. Mechanistically, MST4 inactivation elevates the expression and nuclear translocation of Snail1, a key EMT transcription factor (EMT-TF), through the PI3K/AKT signaling pathway, thus inducing the EMT phenotype of HCC cells, and enhancing their invasive and metastatic potential. Moreover, a negative correlation between MST4 and p-AKT, Snail1, and Ki67 and a positive correlation between MST4 and E-cadherin were determined in clinical HCC samples. Conclusions: Our findings indicate that MST4 suppresses EMT, invasion, and metastasis of HCC cells by modulating the PI3K/AKT/Snail1 axis, suggesting that MST4 may be a potential prognostic biomarker for aggressive and metastatic HCC. Ivyspring International Publisher 2021-05-27 /pmc/articles/PMC8210547/ /pubmed/34149910 http://dx.doi.org/10.7150/jca.60008 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Dian, Mei-Juan
Li, Jing
Zhang, Xiao-Ling
Li, Zi-Jian
Zhou, Ying
Zhou, Wei
Zhong, Qiu-Ling
Pang, Wen-Qian
Lin, Xiao-Lin
Liu, Tao
Liu, Yi-An
Li, Yong-Long
Han, Liu-Xin
Zhao, Wen-Tao
Jia, Jun-Shuang
Xiao, Sheng-Jun
Xiao, Dong
Xia, Jia-Wei
Hao, Wei-Chao
MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title_full MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title_fullStr MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title_full_unstemmed MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title_short MST4 negatively regulates the EMT, invasion and metastasis of HCC cells by inactivating PI3K/AKT/Snail1 axis
title_sort mst4 negatively regulates the emt, invasion and metastasis of hcc cells by inactivating pi3k/akt/snail1 axis
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8210547/
https://www.ncbi.nlm.nih.gov/pubmed/34149910
http://dx.doi.org/10.7150/jca.60008
work_keys_str_mv AT dianmeijuan mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT lijing mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT zhangxiaoling mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT lizijian mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT zhouying mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT zhouwei mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT zhongqiuling mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT pangwenqian mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT linxiaolin mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT liutao mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT liuyian mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT liyonglong mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT hanliuxin mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT zhaowentao mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT jiajunshuang mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT xiaoshengjun mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT xiaodong mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT xiajiawei mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis
AT haoweichao mst4negativelyregulatestheemtinvasionandmetastasisofhcccellsbyinactivatingpi3kaktsnail1axis