树突状细胞中 YTHDF1 的缺失可通过 STING 依赖性 IFN 的产生增强辐射诱导的抗肿瘤免疫。

Chuangyu Wen,Liangliang Wang,András Piffkó,Dapeng Chen,Xianbin Yu,Katarzyna Zawieracz,Jason Bugno,Kaiting Yang,Emile Z Naccasha,Fei Ji,Jiaai Wang,Xiaona Huang,Stephen Y Luo,Lei Tan,Bin Shen,Cheng Luo,Megan E McNerney,Steven J Chmura,Ainhoa Arina,Sean P Pitroda,Chuan He,Hua Liang,Ralph R Weichselbaum
{"title":"树突状细胞中 YTHDF1 的缺失可通过 STING 依赖性 IFN 的产生增强辐射诱导的抗肿瘤免疫。","authors":"Chuangyu Wen,Liangliang Wang,András Piffkó,Dapeng Chen,Xianbin Yu,Katarzyna Zawieracz,Jason Bugno,Kaiting Yang,Emile Z Naccasha,Fei Ji,Jiaai Wang,Xiaona Huang,Stephen Y Luo,Lei Tan,Bin Shen,Cheng Luo,Megan E McNerney,Steven J Chmura,Ainhoa Arina,Sean P Pitroda,Chuan He,Hua Liang,Ralph R Weichselbaum","doi":"10.1172/jci181612","DOIUrl":null,"url":null,"abstract":"RNA N6-methyladenosine (m6A) reader YTHDF1 is implicated in cancer etiology and progression. We discovered that radiotherapy (RT) increased YTHDF1 expression in dendritic cells (DCs) of PBMCs from cancer patients, but not in other immune cells tested. Elevated YTHDF1 expression of DCs was associated with poor outcomes in patients receiving RT. We found that loss of Ythdf1 in DCs enhanced the antitumor effects of ionizing radiation (IR) via increasing the cross-priming capacity of DCs across multiple murine cancer models. Mechanistically, IR upregulated YTHDF1 expression in DCs through STING-IFN-I signaling. YTHDF1 in turn triggered STING degradation by increasing lysosomal cathepsins, thereby reducing IFN-I production. We created a YTHDF1 deletion/inhibition prototype DC vaccine, significantly improving the therapeutic effect of RT and radio-immunotherapy in a murine melanoma model. Our findings reveal a new layer of regulation between YTHDF1/m6A and STING in response to IR, which opens new paths for the development of YTHDF1-targeting therapies.","PeriodicalId":520097,"journal":{"name":"The Journal of Clinical Investigation","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"YTHDF1 loss in dendritic cells potentiates radiation-induced antitumor immunity via STING-dependent type I IFN production.\",\"authors\":\"Chuangyu Wen,Liangliang Wang,András Piffkó,Dapeng Chen,Xianbin Yu,Katarzyna Zawieracz,Jason Bugno,Kaiting Yang,Emile Z Naccasha,Fei Ji,Jiaai Wang,Xiaona Huang,Stephen Y Luo,Lei Tan,Bin Shen,Cheng Luo,Megan E McNerney,Steven J Chmura,Ainhoa Arina,Sean P Pitroda,Chuan He,Hua Liang,Ralph R Weichselbaum\",\"doi\":\"10.1172/jci181612\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"RNA N6-methyladenosine (m6A) reader YTHDF1 is implicated in cancer etiology and progression. We discovered that radiotherapy (RT) increased YTHDF1 expression in dendritic cells (DCs) of PBMCs from cancer patients, but not in other immune cells tested. Elevated YTHDF1 expression of DCs was associated with poor outcomes in patients receiving RT. We found that loss of Ythdf1 in DCs enhanced the antitumor effects of ionizing radiation (IR) via increasing the cross-priming capacity of DCs across multiple murine cancer models. Mechanistically, IR upregulated YTHDF1 expression in DCs through STING-IFN-I signaling. YTHDF1 in turn triggered STING degradation by increasing lysosomal cathepsins, thereby reducing IFN-I production. We created a YTHDF1 deletion/inhibition prototype DC vaccine, significantly improving the therapeutic effect of RT and radio-immunotherapy in a murine melanoma model. Our findings reveal a new layer of regulation between YTHDF1/m6A and STING in response to IR, which opens new paths for the development of YTHDF1-targeting therapies.\",\"PeriodicalId\":520097,\"journal\":{\"name\":\"The Journal of Clinical Investigation\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-09-26\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"The Journal of Clinical Investigation\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1172/jci181612\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"The Journal of Clinical Investigation","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1172/jci181612","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

RNA的N6-甲基腺苷(m6A)阅读器YTHDF1与癌症的病因和进展有关。我们发现,放疗(RT)会增加癌症患者 PBMC 的树突状细胞(DC)中 YTHDF1 的表达,但不会增加其他免疫细胞中 YTHDF1 的表达。DC中YTHDF1表达的升高与接受RT治疗患者的不良预后有关。我们发现,在多种小鼠癌症模型中,DCs 中 Ythdf1 的缺失会通过提高 DCs 的交叉刺激能力来增强电离辐射(IR)的抗肿瘤效果。从机理上讲,IR通过STING-IFN-I信号转导上调DC中YTHDF1的表达。YTHDF1反过来又通过增加溶酶体酪蛋白引发STING降解,从而减少IFN-I的产生。我们创建了一种YTHDF1缺失/抑制原型DC疫苗,显著提高了RT和放射免疫疗法在小鼠黑色素瘤模型中的治疗效果。我们的研究结果揭示了YTHDF1/m6A和STING之间对IR反应的新的调控层,这为开发YTHDF1靶向疗法开辟了新的道路。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
YTHDF1 loss in dendritic cells potentiates radiation-induced antitumor immunity via STING-dependent type I IFN production.
RNA N6-methyladenosine (m6A) reader YTHDF1 is implicated in cancer etiology and progression. We discovered that radiotherapy (RT) increased YTHDF1 expression in dendritic cells (DCs) of PBMCs from cancer patients, but not in other immune cells tested. Elevated YTHDF1 expression of DCs was associated with poor outcomes in patients receiving RT. We found that loss of Ythdf1 in DCs enhanced the antitumor effects of ionizing radiation (IR) via increasing the cross-priming capacity of DCs across multiple murine cancer models. Mechanistically, IR upregulated YTHDF1 expression in DCs through STING-IFN-I signaling. YTHDF1 in turn triggered STING degradation by increasing lysosomal cathepsins, thereby reducing IFN-I production. We created a YTHDF1 deletion/inhibition prototype DC vaccine, significantly improving the therapeutic effect of RT and radio-immunotherapy in a murine melanoma model. Our findings reveal a new layer of regulation between YTHDF1/m6A and STING in response to IR, which opens new paths for the development of YTHDF1-targeting therapies.
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
自引率
0.00%
发文量
0
×
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