CD4+ T 细胞产生 IFN-I,许可 cDC1s 诱导人类肿瘤中的细胞毒性 T 细胞活性

IF 21.8 1区 医学 Q1 IMMUNOLOGY
Xin Lei, Daniël C. de Groot, Marij J. P. Welters, Tom de Wit, Ellen Schrama, Hans van Eenennaam, Saskia J. Santegoets, Timo Oosenbrug, Annemarthe van der Veen, Joris L. Vos, Charlotte L. Zuur, Noel F. C. C. de Miranda, Heinz Jacobs, Sjoerd H. van der Burg, Jannie Borst, Yanling Xiao
{"title":"CD4+ T 细胞产生 IFN-I,许可 cDC1s 诱导人类肿瘤中的细胞毒性 T 细胞活性","authors":"Xin Lei, Daniël C. de Groot, Marij J. P. Welters, Tom de Wit, Ellen Schrama, Hans van Eenennaam, Saskia J. Santegoets, Timo Oosenbrug, Annemarthe van der Veen, Joris L. Vos, Charlotte L. Zuur, Noel F. C. C. de Miranda, Heinz Jacobs, Sjoerd H. van der Burg, Jannie Borst, Yanling Xiao","doi":"10.1038/s41423-024-01133-1","DOIUrl":null,"url":null,"abstract":"CD4+ T cells can \"help” or \"license” conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNβ via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNβ also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver “help” signals to CTLs locally, according to their transcriptomic profile and colocalization with “helped/licensed” cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8000,"publicationDate":"2024-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01133-1.pdf","citationCount":"0","resultStr":"{\"title\":\"CD4+ T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors\",\"authors\":\"Xin Lei, Daniël C. de Groot, Marij J. P. Welters, Tom de Wit, Ellen Schrama, Hans van Eenennaam, Saskia J. Santegoets, Timo Oosenbrug, Annemarthe van der Veen, Joris L. Vos, Charlotte L. Zuur, Noel F. C. C. de Miranda, Heinz Jacobs, Sjoerd H. van der Burg, Jannie Borst, Yanling Xiao\",\"doi\":\"10.1038/s41423-024-01133-1\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"CD4+ T cells can \\\"help” or \\\"license” conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNβ via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNβ also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver “help” signals to CTLs locally, according to their transcriptomic profile and colocalization with “helped/licensed” cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.\",\"PeriodicalId\":9950,\"journal\":{\"name\":\"Cellular &Molecular Immunology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":21.8000,\"publicationDate\":\"2024-02-21\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.nature.com/articles/s41423-024-01133-1.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cellular &Molecular Immunology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.nature.com/articles/s41423-024-01133-1\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cellular &Molecular Immunology","FirstCategoryId":"3","ListUrlMain":"https://www.nature.com/articles/s41423-024-01133-1","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

CD4+ T细胞可以 "帮助 "或 "许可 "传统的1型树突状细胞(cDC1s)诱导CD8+细胞毒性T淋巴细胞(CTL)抗癌反应,这已在小鼠模型中得到证实。我们最近发现了具有 CD4+ T 细胞帮助转录组印记的 cDC1s,特别是在 T 细胞浸润的人类癌症中,这些细胞与良好的预后和对 PD-1 靶向免疫疗法的反应有关。在这里,我们描述了人类 CD4+ T 细胞许可 cDC1 的机制。活化的 CD4+ T 细胞通过 STING 途径产生 IFNβ,促进 cDC1s 的 MHC-I 抗原(交叉)呈递,从而提高它们诱导 CTL 抗癌反应的能力。通过与 CD40 配体(L)合作,IFNβ 还能优化 cDC1s 诱导 CTL 反应所需的成本刺激和其他功能。产生 IFN-I 的 CD4+ T 细胞存在于各种 T 细胞浸润的癌症中,根据它们的转录组特征以及与 "被帮助/被许可 "的 cDC 和肿瘤反应性 CD8+ T 细胞的共定位,它们很可能在局部向 CTL 发出 "帮助 "信号。与这种情况一致的是,TME 中存在产生 IFN-I 的 CD4+ T 细胞与癌症患者的总生存期和对 PD-1 检查点阻断的反应有关。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

CD4+ T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors

CD4+ T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors

CD4+ T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors
CD4+ T cells can "help” or "license” conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNβ via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNβ also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver “help” signals to CTLs locally, according to their transcriptomic profile and colocalization with “helped/licensed” cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
31.20
自引率
1.20%
发文量
903
审稿时长
1 months
期刊介绍: Cellular & Molecular Immunology, a monthly journal from the Chinese Society of Immunology and the University of Science and Technology of China, serves as a comprehensive platform covering both basic immunology research and clinical applications. The journal publishes a variety of article types, including Articles, Review Articles, Mini Reviews, and Short Communications, focusing on diverse aspects of cellular and molecular immunology.
×
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