Targeting a disintegrin and metalloprotease (ADAM) 17-CD122 axis enhances CD8+ T cell effector differentiation and anti-tumor immunity.

IF 40.8 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY
Lina Sun, Anjun Jiao, Haiyan Liu, Renyi Ding, Ning Yuan, Biao Yang, Cangang Zhang, Xiaoxuan Jia, Gang Wang, Yanhong Su, Dan Zhang, Lin Shi, Chenming Sun, Aijun Zhang, Lianjun Zhang, Baojun Zhang
{"title":"Targeting a disintegrin and metalloprotease (ADAM) 17-CD122 axis enhances CD8<sup>+</sup> T cell effector differentiation and anti-tumor immunity.","authors":"Lina Sun, Anjun Jiao, Haiyan Liu, Renyi Ding, Ning Yuan, Biao Yang, Cangang Zhang, Xiaoxuan Jia, Gang Wang, Yanhong Su, Dan Zhang, Lin Shi, Chenming Sun, Aijun Zhang, Lianjun Zhang, Baojun Zhang","doi":"10.1038/s41392-024-01873-6","DOIUrl":null,"url":null,"abstract":"<p><p>CD8<sup>+</sup> T cell immune responses are regulated by multi-layer networks, while the post-translational regulation remains largely unknown. Transmembrane ectodomain shedding is an important post-translational process orchestrating receptor expression and signal transduction through proteolytic cleavage of membrane proteins. Here, by targeting the sheddase A Disintegrin and Metalloprotease (ADAM)17, we defined a post-translational regulatory mechanism mediated by the ectodomain shedding in CD8<sup>+</sup> T cells. Transcriptomic and proteomic analysis revealed the involvement of post-translational regulation in CD8<sup>+</sup> T cells. T cell-specific deletion of ADAM17 led to a dramatic increase in effector CD8<sup>+</sup> T cell differentiation and enhanced cytolytic effects to eliminate pathogens and tumors. Mechanistically, ADAM17 regulated CD8<sup>+</sup> T cells through cleavage of membrane CD122. ADAM17 inhibition led to elevated CD122 expression and enhanced response to IL-2 and IL-15 stimulation in both mouse and human CD8<sup>+</sup> T cells. Intriguingly, inhibition of ADAM17 in CD8<sup>+</sup> T cells improved the efficacy of chimeric antigen receptor (CAR) T cells in solid tumors. Our findings reveal a critical post-translational regulation in CD8<sup>+</sup> T cells, providing a potential therapeutic strategy of targeting ADAM17 for effective anti-tumor immunity.</p>","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":null,"pages":null},"PeriodicalIF":40.8000,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11199508/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Signal Transduction and Targeted Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41392-024-01873-6","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

CD8+ T cell immune responses are regulated by multi-layer networks, while the post-translational regulation remains largely unknown. Transmembrane ectodomain shedding is an important post-translational process orchestrating receptor expression and signal transduction through proteolytic cleavage of membrane proteins. Here, by targeting the sheddase A Disintegrin and Metalloprotease (ADAM)17, we defined a post-translational regulatory mechanism mediated by the ectodomain shedding in CD8+ T cells. Transcriptomic and proteomic analysis revealed the involvement of post-translational regulation in CD8+ T cells. T cell-specific deletion of ADAM17 led to a dramatic increase in effector CD8+ T cell differentiation and enhanced cytolytic effects to eliminate pathogens and tumors. Mechanistically, ADAM17 regulated CD8+ T cells through cleavage of membrane CD122. ADAM17 inhibition led to elevated CD122 expression and enhanced response to IL-2 and IL-15 stimulation in both mouse and human CD8+ T cells. Intriguingly, inhibition of ADAM17 in CD8+ T cells improved the efficacy of chimeric antigen receptor (CAR) T cells in solid tumors. Our findings reveal a critical post-translational regulation in CD8+ T cells, providing a potential therapeutic strategy of targeting ADAM17 for effective anti-tumor immunity.

Abstract Image

靶向崩解素和金属蛋白酶(ADAM)17-CD122 轴可增强 CD8+ T 细胞效应分化和抗肿瘤免疫。
CD8+ T 细胞免疫反应受多层网络调控,而翻译后调控在很大程度上仍是未知的。跨膜外域脱落是一个重要的翻译后过程,它通过膜蛋白的蛋白水解裂解来协调受体表达和信号转导。在这里,通过靶向脱落酶A分解蛋白和金属蛋白酶(ADAM)17,我们定义了一种由CD8+ T细胞外结构域脱落介导的翻译后调控机制。转录组和蛋白质组分析表明,CD8+ T 细胞参与了翻译后调控。T细胞特异性地缺失ADAM17会导致效应CD8+ T细胞的分化急剧增加,并增强细胞溶解效应以消灭病原体和肿瘤。从机制上讲,ADAM17 通过裂解膜 CD122 来调节 CD8+ T 细胞。抑制ADAM17会导致CD122表达升高,并增强小鼠和人类CD8+ T细胞对IL-2和IL-15刺激的反应。有趣的是,抑制 CD8+ T 细胞中的 ADAM17 能提高嵌合抗原受体(CAR)T 细胞在实体瘤中的疗效。我们的研究结果揭示了 CD8+ T 细胞中一个关键的翻译后调控机制,为靶向 ADAM17 以实现有效的抗肿瘤免疫提供了一种潜在的治疗策略。
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
Signal Transduction and Targeted Therapy
Signal Transduction and Targeted Therapy Biochemistry, Genetics and Molecular Biology-Genetics
CiteScore
44.50
自引率
1.50%
发文量
384
审稿时长
5 weeks
期刊介绍: Signal Transduction and Targeted Therapy is an open access journal that focuses on timely publication of cutting-edge discoveries and advancements in basic science and clinical research related to signal transduction and targeted therapy. Scope: The journal covers research on major human diseases, including, but not limited to: Cancer,Cardiovascular diseases,Autoimmune diseases,Nervous system diseases.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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