Cargando…

Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation

The thioredoxin reductase (TrxR) 1 is often overexpressed in numerous cancer cells. Targeting TrxR1 leads to a reduction in tumor progression and metastasis, making the enzyme an attractive target for cancer treatment. Our previous research revealed that the curcumin derivative B19 could induce canc...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Weiqian, Zou, Peng, Zhao, Zhongwei, Weng, Qiaoyou, Chen, Xi, Ying, Shilong, Ye, Qingqing, Wang, Zhe, Ji, Jiansong, Liang, Guang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4941337/
https://www.ncbi.nlm.nih.gov/pubmed/26919094
http://dx.doi.org/10.18632/oncotarget.7565
_version_ 1782442289130373120
author Chen, Weiqian
Zou, Peng
Zhao, Zhongwei
Weng, Qiaoyou
Chen, Xi
Ying, Shilong
Ye, Qingqing
Wang, Zhe
Ji, Jiansong
Liang, Guang
author_facet Chen, Weiqian
Zou, Peng
Zhao, Zhongwei
Weng, Qiaoyou
Chen, Xi
Ying, Shilong
Ye, Qingqing
Wang, Zhe
Ji, Jiansong
Liang, Guang
author_sort Chen, Weiqian
collection PubMed
description The thioredoxin reductase (TrxR) 1 is often overexpressed in numerous cancer cells. Targeting TrxR1 leads to a reduction in tumor progression and metastasis, making the enzyme an attractive target for cancer treatment. Our previous research revealed that the curcumin derivative B19 could induce cancer cell apoptosis via activation of endoplasmic reticulum (ER) stress. However, the upstream mechanism and molecular target of B19 is still unclear. In this study, we demonstrate that B19 directly inhibits TrxR1 enzyme activity to elevate oxidative stress and then induce ROS-mediated ER Stress and mitochondrial dysfunction, subsequently resulting in cell cycle arrest and apoptosis in human gastric cancer cells. A computer-assistant docking showed that B19 may bind TrxR1 protein via formation of a covalent bond with the residue Cys-498. Blockage of ROS production totally reversed B19-induced anti-cancer actions. In addition, the results of xenograft experiments in mice were highly consistent with in vitro studies. Taken together, targeting TrxR1 with B19 provides deep insight into the understanding of how B19 exerts its anticancer effects. More importantly, this work indicates that targeting TrxR1 and manipulating ROS levels are effective therapeutic strategy for the treatment of gastric cancer.
format Online
Article
Text
id pubmed-4941337
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-49413372016-07-19 Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation Chen, Weiqian Zou, Peng Zhao, Zhongwei Weng, Qiaoyou Chen, Xi Ying, Shilong Ye, Qingqing Wang, Zhe Ji, Jiansong Liang, Guang Oncotarget Research Paper The thioredoxin reductase (TrxR) 1 is often overexpressed in numerous cancer cells. Targeting TrxR1 leads to a reduction in tumor progression and metastasis, making the enzyme an attractive target for cancer treatment. Our previous research revealed that the curcumin derivative B19 could induce cancer cell apoptosis via activation of endoplasmic reticulum (ER) stress. However, the upstream mechanism and molecular target of B19 is still unclear. In this study, we demonstrate that B19 directly inhibits TrxR1 enzyme activity to elevate oxidative stress and then induce ROS-mediated ER Stress and mitochondrial dysfunction, subsequently resulting in cell cycle arrest and apoptosis in human gastric cancer cells. A computer-assistant docking showed that B19 may bind TrxR1 protein via formation of a covalent bond with the residue Cys-498. Blockage of ROS production totally reversed B19-induced anti-cancer actions. In addition, the results of xenograft experiments in mice were highly consistent with in vitro studies. Taken together, targeting TrxR1 with B19 provides deep insight into the understanding of how B19 exerts its anticancer effects. More importantly, this work indicates that targeting TrxR1 and manipulating ROS levels are effective therapeutic strategy for the treatment of gastric cancer. Impact Journals LLC 2016-02-22 /pmc/articles/PMC4941337/ /pubmed/26919094 http://dx.doi.org/10.18632/oncotarget.7565 Text en Copyright: © 2016 Chen et al. http://creativecommons.org/licenses/by/2.5/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Chen, Weiqian
Zou, Peng
Zhao, Zhongwei
Weng, Qiaoyou
Chen, Xi
Ying, Shilong
Ye, Qingqing
Wang, Zhe
Ji, Jiansong
Liang, Guang
Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title_full Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title_fullStr Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title_full_unstemmed Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title_short Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation
title_sort selective killing of gastric cancer cells by a small molecule via targeting trxr1 and ros-mediated er stress activation
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4941337/
https://www.ncbi.nlm.nih.gov/pubmed/26919094
http://dx.doi.org/10.18632/oncotarget.7565
work_keys_str_mv AT chenweiqian selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT zoupeng selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT zhaozhongwei selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT wengqiaoyou selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT chenxi selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT yingshilong selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT yeqingqing selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT wangzhe selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT jijiansong selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation
AT liangguang selectivekillingofgastriccancercellsbyasmallmoleculeviatargetingtrxr1androsmediatederstressactivation