IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer

Ferroptosis, implicated in several diseases, is a new form of programmed and nonapoptotic cell death triggered by iron-dependent lipid peroxidation after inactivation of the cystine/glutamate antiporter system xc–, which is composed of solute carrier family 7 membrane 11 (SLC7A11) and solute carrier...

Full description

Bibliographic Details
Main Authors: Lei Zhang, Wen Liu, Fangyan Liu, Qun Wang, Mengjiao Song, Qi Yu, Kun Tang, Tieshan Teng, Dongdong Wu, Xijing Wang, Wuqi Han, Yanzhang Li
Format: Article
Language:English
Published: Hindawi Limited 2020-01-01
Series:Oxidative Medicine and Cellular Longevity
Online Access:http://dx.doi.org/10.1155/2020/1675613
id doaj-d26945723d5e4da4b0f2463c9b3afdb2
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Lei Zhang
Wen Liu
Fangyan Liu
Qun Wang
Mengjiao Song
Qi Yu
Kun Tang
Tieshan Teng
Dongdong Wu
Xijing Wang
Wuqi Han
Yanzhang Li
spellingShingle Lei Zhang
Wen Liu
Fangyan Liu
Qun Wang
Mengjiao Song
Qi Yu
Kun Tang
Tieshan Teng
Dongdong Wu
Xijing Wang
Wuqi Han
Yanzhang Li
IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
Oxidative Medicine and Cellular Longevity
author_facet Lei Zhang
Wen Liu
Fangyan Liu
Qun Wang
Mengjiao Song
Qi Yu
Kun Tang
Tieshan Teng
Dongdong Wu
Xijing Wang
Wuqi Han
Yanzhang Li
author_sort Lei Zhang
title IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
title_short IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
title_full IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
title_fullStr IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
title_full_unstemmed IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer
title_sort imca induces ferroptosis mediated by slc7a11 through the ampk/mtor pathway in colorectal cancer
publisher Hindawi Limited
series Oxidative Medicine and Cellular Longevity
issn 1942-0900
1942-0994
publishDate 2020-01-01
description Ferroptosis, implicated in several diseases, is a new form of programmed and nonapoptotic cell death triggered by iron-dependent lipid peroxidation after inactivation of the cystine/glutamate antiporter system xc–, which is composed of solute carrier family 7 membrane 11 (SLC7A11) and solute carrier family 3 membrane 2 (SLC3A2). Therefore, inducing ferroptosis through inhibiting the cystine/glutamate antiporter system xc– may be an effective way to treat cancer. In previous screening tests, we found that the benzopyran derivative 2-imino-6-methoxy-2H-chromene-3-carbothioamide (IMCA) significantly inhibited the viability of colorectal cancer cells. However, the impact of IMCA on ferroptosis remains unknown. Hence, this study investigated the effect of IMCA on ferroptosis and elucidated the underlying molecular mechanism. Results showed that IMCA significantly inhibited the cell viability of colorectal cancer cells in vitro and inhibited tumor growth with negligible organ toxicity in vivo. Further studies showed that IMCA significantly induced the ferroptosis of colorectal cancer cells. Mechanistically, IMCA downregulated the expression of SLC7A11 and decreased the contents of cysteine and glutathione, which resulted in reactive oxygen species accumulation and ferroptosis. Furthermore, overexpression of SLC7A11 significantly attenuated the ferroptosis caused by IMCA. In addition, IMCA regulated the activity of the AMPK/mTOR/p70S6k signaling pathway, which is related to the activity of SLC7A11 and ferroptosis. Collectively, our research provided experimental evidences on the activity and mechanism of ferroptosis induced by IMCA and revealed that IMCA might be a promising therapeutic drug for colorectal cancer.
url http://dx.doi.org/10.1155/2020/1675613
work_keys_str_mv AT leizhang imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT wenliu imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT fangyanliu imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT qunwang imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT mengjiaosong imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT qiyu imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT kuntang imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT tieshanteng imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT dongdongwu imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT xijingwang imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT wuqihan imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
AT yanzhangli imcainducesferroptosismediatedbyslc7a11throughtheampkmtorpathwayincolorectalcancer
_version_ 1715582556029583360
spelling doaj-d26945723d5e4da4b0f2463c9b3afdb22020-11-25T02:04:12ZengHindawi LimitedOxidative Medicine and Cellular Longevity1942-09001942-09942020-01-01202010.1155/2020/16756131675613IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal CancerLei Zhang0Wen Liu1Fangyan Liu2Qun Wang3Mengjiao Song4Qi Yu5Kun Tang6Tieshan Teng7Dongdong Wu8Xijing Wang9Wuqi Han10Yanzhang Li11Cell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaDepartment of Dermatology, Second People’s Hospital of Zhengzhou, Zhengzhou 450006, ChinaKaifeng Food and Drug Inspection Institute, Kaifeng 475004, ChinaCell Signal Transduction Laboratory, Bioinformatics Center, Laboratory for Nanomedicine, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, ChinaFerroptosis, implicated in several diseases, is a new form of programmed and nonapoptotic cell death triggered by iron-dependent lipid peroxidation after inactivation of the cystine/glutamate antiporter system xc–, which is composed of solute carrier family 7 membrane 11 (SLC7A11) and solute carrier family 3 membrane 2 (SLC3A2). Therefore, inducing ferroptosis through inhibiting the cystine/glutamate antiporter system xc– may be an effective way to treat cancer. In previous screening tests, we found that the benzopyran derivative 2-imino-6-methoxy-2H-chromene-3-carbothioamide (IMCA) significantly inhibited the viability of colorectal cancer cells. However, the impact of IMCA on ferroptosis remains unknown. Hence, this study investigated the effect of IMCA on ferroptosis and elucidated the underlying molecular mechanism. Results showed that IMCA significantly inhibited the cell viability of colorectal cancer cells in vitro and inhibited tumor growth with negligible organ toxicity in vivo. Further studies showed that IMCA significantly induced the ferroptosis of colorectal cancer cells. Mechanistically, IMCA downregulated the expression of SLC7A11 and decreased the contents of cysteine and glutathione, which resulted in reactive oxygen species accumulation and ferroptosis. Furthermore, overexpression of SLC7A11 significantly attenuated the ferroptosis caused by IMCA. In addition, IMCA regulated the activity of the AMPK/mTOR/p70S6k signaling pathway, which is related to the activity of SLC7A11 and ferroptosis. Collectively, our research provided experimental evidences on the activity and mechanism of ferroptosis induced by IMCA and revealed that IMCA might be a promising therapeutic drug for colorectal cancer.http://dx.doi.org/10.1155/2020/1675613