Iron-loaded cancer-associated fibroblasts induce immunosuppression in prostate cancer

IF 14.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES
Kai Zhang, Kaiyuan Liu, Benxia Hu, Genyu Du, Xinyu Chen, Lingling Xiao, Yingchao Zhang, Luyao Jiang, Na Jing, Chaping Cheng, Jinming Wang, Penghui Xu, You Wang, Pengfei Ma, Guanglei Zhuang, Huifang Zhao, Yujiao Sun, Deng Wang, Qi Wang, Wei Xue, Wei-Qiang Gao, Pengcheng Zhang, Helen He Zhu
{"title":"Iron-loaded cancer-associated fibroblasts induce immunosuppression in prostate cancer","authors":"Kai Zhang, Kaiyuan Liu, Benxia Hu, Genyu Du, Xinyu Chen, Lingling Xiao, Yingchao Zhang, Luyao Jiang, Na Jing, Chaping Cheng, Jinming Wang, Penghui Xu, You Wang, Pengfei Ma, Guanglei Zhuang, Huifang Zhao, Yujiao Sun, Deng Wang, Qi Wang, Wei Xue, Wei-Qiang Gao, Pengcheng Zhang, Helen He Zhu","doi":"10.1038/s41467-024-53233-1","DOIUrl":null,"url":null,"abstract":"<p>Iron is an essential biomineral in the human body. Here, we describe a subset of iron-loaded cancer-associated fibroblasts, termed as FerroCAFs, that utilize iron to induce immunosuppression in prostate cancer and predict an unfavorable clinical outcome. FerroCAFs secrete myeloid cell-associated proteins, including CCL2, CSF1 and CXCL1, to recruit immunosuppressive myeloid cells. We report the presence of FerroCAFs in prostate cancer from both mice and human, as well as in human lung and ovarian cancers, and identify a conserved cell surface marker, the poliovirus receptor. Mechanistically, the accumulated iron in FerroCAFs is caused by Hmox1-mediated iron release from heme degradation. The intracellular iron activates the Kdm6b, an iron-dependent epigenetic enzyme, to induce an accessible chromatin state and transcription of myeloid cell-associated protein genes. Targeting the FerroCAFs by inhibiting the Hmox1/iron/Kdm6b signaling axis incurs anti-tumor immunity and tumor suppression. Collectively, we report an iron-loaded FerroCAF cluster that drives immunosuppression through an iron-dependent epigenetic reprogramming mechanism and reveal promising therapeutic targets to boost anti-tumor immunity.</p>","PeriodicalId":19066,"journal":{"name":"Nature Communications","volume":null,"pages":null},"PeriodicalIF":14.7000,"publicationDate":"2024-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Nature Communications","FirstCategoryId":"103","ListUrlMain":"https://doi.org/10.1038/s41467-024-53233-1","RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MULTIDISCIPLINARY SCIENCES","Score":null,"Total":0}
引用次数: 0

Abstract

Iron is an essential biomineral in the human body. Here, we describe a subset of iron-loaded cancer-associated fibroblasts, termed as FerroCAFs, that utilize iron to induce immunosuppression in prostate cancer and predict an unfavorable clinical outcome. FerroCAFs secrete myeloid cell-associated proteins, including CCL2, CSF1 and CXCL1, to recruit immunosuppressive myeloid cells. We report the presence of FerroCAFs in prostate cancer from both mice and human, as well as in human lung and ovarian cancers, and identify a conserved cell surface marker, the poliovirus receptor. Mechanistically, the accumulated iron in FerroCAFs is caused by Hmox1-mediated iron release from heme degradation. The intracellular iron activates the Kdm6b, an iron-dependent epigenetic enzyme, to induce an accessible chromatin state and transcription of myeloid cell-associated protein genes. Targeting the FerroCAFs by inhibiting the Hmox1/iron/Kdm6b signaling axis incurs anti-tumor immunity and tumor suppression. Collectively, we report an iron-loaded FerroCAF cluster that drives immunosuppression through an iron-dependent epigenetic reprogramming mechanism and reveal promising therapeutic targets to boost anti-tumor immunity.

Abstract Image

铁负载癌症相关成纤维细胞诱导前列腺癌免疫抑制
铁是人体必需的生物矿物质。在这里,我们描述了一种铁负荷的癌症相关成纤维细胞亚群,称为铁前列腺癌相关成纤维细胞(FerroCAFs),它们利用铁诱导前列腺癌的免疫抑制,并预测不利的临床结果。FerroCAFs会分泌髓系细胞相关蛋白,包括CCL2、CSF1和CXCL1,以招募免疫抑制性髓系细胞。我们报告了小鼠和人类前列腺癌以及人类肺癌和卵巢癌中存在铁CAF,并确定了一种保守的细胞表面标记--脊髓灰质炎病毒受体。从机理上讲,FerroCAFs 中积累的铁是由 Hmox1 介导的血红素降解释放铁造成的。细胞内的铁激活了铁依赖性表观遗传酶 Kdm6b,从而诱导染色质状态和髓细胞相关蛋白基因的转录。通过抑制 Hmox1/铁/Kdm6b 信号轴来靶向铁CAFs,可产生抗肿瘤免疫和抑制肿瘤的作用。总之,我们报告了一种通过铁依赖的表观遗传学重编程机制驱动免疫抑制的铁负荷铁CAF簇,并揭示了增强抗肿瘤免疫力的有希望的治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Nature Communications
Nature Communications Biological Science Disciplines-
CiteScore
24.90
自引率
2.40%
发文量
6928
审稿时长
3.7 months
期刊介绍: Nature Communications, an open-access journal, publishes high-quality research spanning all areas of the natural sciences. Papers featured in the journal showcase significant advances relevant to specialists in each respective field. With a 2-year impact factor of 16.6 (2022) and a median time of 8 days from submission to the first editorial decision, Nature Communications is committed to rapid dissemination of research findings. As a multidisciplinary journal, it welcomes contributions from biological, health, physical, chemical, Earth, social, mathematical, applied, and engineering sciences, aiming to highlight important breakthroughs within each domain.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信