Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation

As the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cance...

Full description

Bibliographic Details
Main Authors: Lizhong Yao, Junyi Hou, Xiongyan Wu, Yifan Lu, Zhijian Jin, Zhenjia Yu, Beiqin Yu, Jianfang Li, Zhongyin Yang, Chen Li, Min Yan, Zhenggang Zhu, Bingya Liu, Chao Yan, Liping Su
Format: Article
Language:English
Published: Elsevier 2023-11-01
Series:Redox Biology
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S2213231723003245
_version_ 1797647868366946304
author Lizhong Yao
Junyi Hou
Xiongyan Wu
Yifan Lu
Zhijian Jin
Zhenjia Yu
Beiqin Yu
Jianfang Li
Zhongyin Yang
Chen Li
Min Yan
Zhenggang Zhu
Bingya Liu
Chao Yan
Liping Su
author_facet Lizhong Yao
Junyi Hou
Xiongyan Wu
Yifan Lu
Zhijian Jin
Zhenjia Yu
Beiqin Yu
Jianfang Li
Zhongyin Yang
Chen Li
Min Yan
Zhenggang Zhu
Bingya Liu
Chao Yan
Liping Su
author_sort Lizhong Yao
collection DOAJ
description As the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cancer (GC) remain poorly understood. In this study, we demonstrate that NK cell levels are inversely correlated with CAFs abundance in human GC. CAFs impair the anti-tumor capacity of NK cells by inducing ferroptosis, a cell death process characterized by the accumulation of iron-dependent lipid peroxides. CAFs induce ferroptosis in NK cells by promoting iron overload; conversely, decreased intracellular iron levels protect NK cells against CAF-induced ferroptosis. Mechanistically, CAFs increase the labile iron pool within NK cells via iron export into the TME, which is mediated by the upregulated expression of iron regulatory genes ferroportin1 and hephaestin in CAFs. Moreover, CAF-derived follistatin like protein 1(FSTL1) upregulates NCOA4 expression in NK cells via the DIP2A-P38 pathway, and NCOA4-mediated ferritinophagy is required for CAF-induced NK cell ferroptosis. In a human patient-derived organoid model, functional targeting of CAFs using a combination of deferoxamine and FSTL1-neutralizing antibody significantly alleviate CAF-induced NK cell ferroptosis and boost the cytotoxicity of NK cells against GC. This study demonstrates a novel mechanism of suppression of NK cell activity by CAFs in the TME and presents a potential therapeutic approach to augment the immune response against GC mediated by NK cells.
first_indexed 2024-03-11T15:22:51Z
format Article
id doaj.art-1a8ad35e533843c1aa456774f9853db2
institution Directory Open Access Journal
issn 2213-2317
language English
last_indexed 2024-03-11T15:22:51Z
publishDate 2023-11-01
publisher Elsevier
record_format Article
series Redox Biology
spelling doaj.art-1a8ad35e533843c1aa456774f9853db22023-10-28T05:07:19ZengElsevierRedox Biology2213-23172023-11-0167102923Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulationLizhong Yao0Junyi Hou1Xiongyan Wu2Yifan Lu3Zhijian Jin4Zhenjia Yu5Beiqin Yu6Jianfang Li7Zhongyin Yang8Chen Li9Min Yan10Zhenggang Zhu11Bingya Liu12Chao Yan13Liping Su14Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaDepartment of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaCorresponding author.; Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaCorresponding author.; Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, ChinaAs the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cancer (GC) remain poorly understood. In this study, we demonstrate that NK cell levels are inversely correlated with CAFs abundance in human GC. CAFs impair the anti-tumor capacity of NK cells by inducing ferroptosis, a cell death process characterized by the accumulation of iron-dependent lipid peroxides. CAFs induce ferroptosis in NK cells by promoting iron overload; conversely, decreased intracellular iron levels protect NK cells against CAF-induced ferroptosis. Mechanistically, CAFs increase the labile iron pool within NK cells via iron export into the TME, which is mediated by the upregulated expression of iron regulatory genes ferroportin1 and hephaestin in CAFs. Moreover, CAF-derived follistatin like protein 1(FSTL1) upregulates NCOA4 expression in NK cells via the DIP2A-P38 pathway, and NCOA4-mediated ferritinophagy is required for CAF-induced NK cell ferroptosis. In a human patient-derived organoid model, functional targeting of CAFs using a combination of deferoxamine and FSTL1-neutralizing antibody significantly alleviate CAF-induced NK cell ferroptosis and boost the cytotoxicity of NK cells against GC. This study demonstrates a novel mechanism of suppression of NK cell activity by CAFs in the TME and presents a potential therapeutic approach to augment the immune response against GC mediated by NK cells.http://www.sciencedirect.com/science/article/pii/S2213231723003245Cancer-associated fibroblastsNK cellsIron regulationFerroptosisFerritinophagy
spellingShingle Lizhong Yao
Junyi Hou
Xiongyan Wu
Yifan Lu
Zhijian Jin
Zhenjia Yu
Beiqin Yu
Jianfang Li
Zhongyin Yang
Chen Li
Min Yan
Zhenggang Zhu
Bingya Liu
Chao Yan
Liping Su
Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
Redox Biology
Cancer-associated fibroblasts
NK cells
Iron regulation
Ferroptosis
Ferritinophagy
title Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
title_full Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
title_fullStr Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
title_full_unstemmed Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
title_short Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation
title_sort cancer associated fibroblasts impair the cytotoxic function of nk cells in gastric cancer by inducing ferroptosis via iron regulation
topic Cancer-associated fibroblasts
NK cells
Iron regulation
Ferroptosis
Ferritinophagy
url http://www.sciencedirect.com/science/article/pii/S2213231723003245
work_keys_str_mv AT lizhongyao cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT junyihou cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT xiongyanwu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT yifanlu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT zhijianjin cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT zhenjiayu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT beiqinyu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT jianfangli cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT zhongyinyang cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT chenli cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT minyan cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT zhenggangzhu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT bingyaliu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT chaoyan cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation
AT lipingsu cancerassociatedfibroblastsimpairthecytotoxicfunctionofnkcellsingastriccancerbyinducingferroptosisviaironregulation