Cargando…

TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell

Chronic inflammation-induced metastases have long been regarded as one of the significant obstacles in treating cancer. Tumor necrosis factor-α (TNF-α), a main inflammation mediator within tumor microenvironment, affects tumor development by inducing multiple chemokines to establish a complex networ...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Zhengcheng, Ao, Xiang, Shen, Zhilin, Ao, Luoquan, Wu, Xiaofeng, Pu, Chengxiu, Guo, Wei, Xing, Wei, He, Min, Yuan, Hongfeng, Yu, Jianhua, Li, Ling, Xu, Xiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8326125/
https://www.ncbi.nlm.nih.gov/pubmed/34345201
http://dx.doi.org/10.7150/ijbs.61350
_version_ 1783731707251261440
author Wang, Zhengcheng
Ao, Xiang
Shen, Zhilin
Ao, Luoquan
Wu, Xiaofeng
Pu, Chengxiu
Guo, Wei
Xing, Wei
He, Min
Yuan, Hongfeng
Yu, Jianhua
Li, Ling
Xu, Xiang
author_facet Wang, Zhengcheng
Ao, Xiang
Shen, Zhilin
Ao, Luoquan
Wu, Xiaofeng
Pu, Chengxiu
Guo, Wei
Xing, Wei
He, Min
Yuan, Hongfeng
Yu, Jianhua
Li, Ling
Xu, Xiang
author_sort Wang, Zhengcheng
collection PubMed
description Chronic inflammation-induced metastases have long been regarded as one of the significant obstacles in treating cancer. Tumor necrosis factor-α (TNF-α), a main inflammation mediator within tumor microenvironment, affects tumor development by inducing multiple chemokines to establish a complex network. Recent reports have revealed that CXCL10/CXCR3 axis affects cancer cells invasiveness and metastases, and Epithelial-mesenchymal transition (EMT) is the main reason for frequent proliferation and distant organ metastases of colon cancer (CC) cells, However, it is unclear whether TNF-α- mediated chronic inflammation can synergically enhance EMT-mediated CC metastasis through promoting chemokine expression. According to this study, TNF-α activated the PI3K/Akt and p38 MAPK parallel signal transduction pathways, then stimulate downstream NF-κB pathway p65 into the nucleus to activate CXCL10 transcription. CXCL10 enhanced the metastases of CC-cells by triggering small GTPases such as RhoA and cdc42. Furthermore, overexpression of CXCL10 significantly enhanced tumorigenicity and mobility of CC cells in vivo. We further clarified that CXCL10 activated the PI3K/Akt pathway through CXCR3, resulting in suppression of GSK-3β phosphorylation and leading to upregulation of Snail expression, thereby regulating EMT in CC cells. These outcomes lay the foundation for finding new targets to inhibit CC metastases.
format Online
Article
Text
id pubmed-8326125
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-83261252021-08-02 TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell Wang, Zhengcheng Ao, Xiang Shen, Zhilin Ao, Luoquan Wu, Xiaofeng Pu, Chengxiu Guo, Wei Xing, Wei He, Min Yuan, Hongfeng Yu, Jianhua Li, Ling Xu, Xiang Int J Biol Sci Research Paper Chronic inflammation-induced metastases have long been regarded as one of the significant obstacles in treating cancer. Tumor necrosis factor-α (TNF-α), a main inflammation mediator within tumor microenvironment, affects tumor development by inducing multiple chemokines to establish a complex network. Recent reports have revealed that CXCL10/CXCR3 axis affects cancer cells invasiveness and metastases, and Epithelial-mesenchymal transition (EMT) is the main reason for frequent proliferation and distant organ metastases of colon cancer (CC) cells, However, it is unclear whether TNF-α- mediated chronic inflammation can synergically enhance EMT-mediated CC metastasis through promoting chemokine expression. According to this study, TNF-α activated the PI3K/Akt and p38 MAPK parallel signal transduction pathways, then stimulate downstream NF-κB pathway p65 into the nucleus to activate CXCL10 transcription. CXCL10 enhanced the metastases of CC-cells by triggering small GTPases such as RhoA and cdc42. Furthermore, overexpression of CXCL10 significantly enhanced tumorigenicity and mobility of CC cells in vivo. We further clarified that CXCL10 activated the PI3K/Akt pathway through CXCR3, resulting in suppression of GSK-3β phosphorylation and leading to upregulation of Snail expression, thereby regulating EMT in CC cells. These outcomes lay the foundation for finding new targets to inhibit CC metastases. Ivyspring International Publisher 2021-06-26 /pmc/articles/PMC8326125/ /pubmed/34345201 http://dx.doi.org/10.7150/ijbs.61350 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
Wang, Zhengcheng
Ao, Xiang
Shen, Zhilin
Ao, Luoquan
Wu, Xiaofeng
Pu, Chengxiu
Guo, Wei
Xing, Wei
He, Min
Yuan, Hongfeng
Yu, Jianhua
Li, Ling
Xu, Xiang
TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title_full TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title_fullStr TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title_full_unstemmed TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title_short TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell
title_sort tnf-α augments cxcl10/cxcr3 axis activity to induce epithelial-mesenchymal transition in colon cancer cell
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8326125/
https://www.ncbi.nlm.nih.gov/pubmed/34345201
http://dx.doi.org/10.7150/ijbs.61350
work_keys_str_mv AT wangzhengcheng tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT aoxiang tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT shenzhilin tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT aoluoquan tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT wuxiaofeng tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT puchengxiu tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT guowei tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT xingwei tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT hemin tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT yuanhongfeng tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT yujianhua tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT liling tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell
AT xuxiang tnfaaugmentscxcl10cxcr3axisactivitytoinduceepithelialmesenchymaltransitionincoloncancercell