Intestinal epithelial damage-derived mtDNA activates STING-IL12 axis in dendritic cells to promote colitis.

IF 12.4 1区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
Theranostics Pub Date : 2024-07-16 eCollection Date: 2024-01-01 DOI:10.7150/thno.96184
Yajie Cai, Shuo Li, Yang Yang, Shuni Duan, Guifang Fan, Jinzhao Bai, Qi Zheng, Yiqing Gu, Xiaojiaoyang Li, Runping Liu
{"title":"Intestinal epithelial damage-derived mtDNA activates STING-IL12 axis in dendritic cells to promote colitis.","authors":"Yajie Cai, Shuo Li, Yang Yang, Shuni Duan, Guifang Fan, Jinzhao Bai, Qi Zheng, Yiqing Gu, Xiaojiaoyang Li, Runping Liu","doi":"10.7150/thno.96184","DOIUrl":null,"url":null,"abstract":"<p><p><b>Rationale:</b> The treatment of ulcerative colitis (UC) presents an ongoing clinical challenge. Emerging research has implicated that the cGAS-STING pathway promotes the progression of UC, but conflicting results have hindered the development of STING as a therapeutic target. In the current study, we aim to comprehensively elucidate the origins, downstream signaling and pathogenic roles of myeloid STING in colitis and colitis-associated carcinoma (CAC). <b>Methods:</b> <i>Tmem173</i> <sup>fl/fl</sup> <i>Lyz2-Cre</i> <sup>ert2</sup> mice were constructed for inducible myeloid-specific deletion of STING. RNA-sequencing, flow cytometry, and multiplex immunohistochemistry were employed to investigate immune responses in DSS-induced colitis or AOM/DSS-induced carcinogenesis. Colonic organoids, primary bone marrow derived macrophages and dendritic cells, and splenic T cells were used for <i>in vitro</i> studies. <b>Results:</b> We observed that myeloid STING knockout in adult mice inhibited macrophage maturation, reduced DC cell activation, and suppressed pro-inflammatory Th1 and Th17 cells, thereby protecting against both acute and chronic colitis and CAC. However, myeloid STING deletion in neonatal or tumor-present mice exhibited impaired immune tolerance and anti-tumor immunity. Furthermore, we found that TFAM-associated mtDNA released from damaged colonic organoids, rather than bacterial products, activates STING in dendritic cells in an extracellular vesicle-independent yet endocytosis-dependent manner. Both IRF3 and NF-κB are required for STING-mediated expression of IL-12 family cytokines, promoting Th1 and Th17 differentiation and contributing to excessive inflammation in colitis. <b>Conclusions:</b> Detection of the TFAM-mtDNA complex from damaged intestinal epithelium by myeloid STING exacerbates colitis through IL-12 cytokines, providing new evidence to support the development of STING as a therapeutic target for UC and CAC.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":null,"pages":null},"PeriodicalIF":12.4000,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11303083/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Theranostics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/thno.96184","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Rationale: The treatment of ulcerative colitis (UC) presents an ongoing clinical challenge. Emerging research has implicated that the cGAS-STING pathway promotes the progression of UC, but conflicting results have hindered the development of STING as a therapeutic target. In the current study, we aim to comprehensively elucidate the origins, downstream signaling and pathogenic roles of myeloid STING in colitis and colitis-associated carcinoma (CAC). Methods: Tmem173 fl/fl Lyz2-Cre ert2 mice were constructed for inducible myeloid-specific deletion of STING. RNA-sequencing, flow cytometry, and multiplex immunohistochemistry were employed to investigate immune responses in DSS-induced colitis or AOM/DSS-induced carcinogenesis. Colonic organoids, primary bone marrow derived macrophages and dendritic cells, and splenic T cells were used for in vitro studies. Results: We observed that myeloid STING knockout in adult mice inhibited macrophage maturation, reduced DC cell activation, and suppressed pro-inflammatory Th1 and Th17 cells, thereby protecting against both acute and chronic colitis and CAC. However, myeloid STING deletion in neonatal or tumor-present mice exhibited impaired immune tolerance and anti-tumor immunity. Furthermore, we found that TFAM-associated mtDNA released from damaged colonic organoids, rather than bacterial products, activates STING in dendritic cells in an extracellular vesicle-independent yet endocytosis-dependent manner. Both IRF3 and NF-κB are required for STING-mediated expression of IL-12 family cytokines, promoting Th1 and Th17 differentiation and contributing to excessive inflammation in colitis. Conclusions: Detection of the TFAM-mtDNA complex from damaged intestinal epithelium by myeloid STING exacerbates colitis through IL-12 cytokines, providing new evidence to support the development of STING as a therapeutic target for UC and CAC.

肠上皮损伤衍生的 mtDNA 可激活树突状细胞中的 STING-IL12 轴,从而促进结肠炎。
理由:溃疡性结肠炎(UC)的治疗一直是临床难题。新近的研究表明,cGAS-STING 通路会促进 UC 的恶化,但相互矛盾的结果阻碍了 STING 作为治疗靶点的发展。在本研究中,我们旨在全面阐明髓系 STING 在结肠炎和结肠炎相关癌(CAC)中的起源、下游信号转导和致病作用。研究方法构建Tmem173 fl/fl Lyz2-Cre ert2小鼠,诱导髓系特异性缺失STING。采用 RNA 序列测定法、流式细胞术和多重免疫组织化学法研究 DSS 诱导的结肠炎或 AOM/DSS 诱导的癌变中的免疫反应。体外研究使用了结肠器官组织、原始骨髓衍生巨噬细胞和树突状细胞以及脾脏 T 细胞。研究结果我们观察到,成年小鼠髓系 STING 基因敲除可抑制巨噬细胞成熟,减少 DC 细胞活化,抑制促炎性 Th1 和 Th17 细胞,从而防止急性和慢性结肠炎以及 CAC 的发生。然而,在新生小鼠或存在肿瘤的小鼠中,骨髓 STING 缺失会损害免疫耐受和抗肿瘤免疫。此外,我们还发现,从受损结肠器官组织中释放的 TFAM 相关 mtDNA(而非细菌产物)能以不依赖细胞外囊泡但依赖内吞的方式激活树突状细胞中的 STING。STING 介导的 IL-12 家族细胞因子的表达需要 IRF3 和 NF-κB,它们可促进 Th1 和 Th17 分化,并导致结肠炎的过度炎症。结论髓系 STING 检测受损肠上皮的 TFAM-mtDNA 复合物会通过 IL-12 细胞因子加剧结肠炎,这为 STING 成为 UC 和 CAC 的治疗靶点提供了新的证据。
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
Theranostics
Theranostics MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
25.40
自引率
1.60%
发文量
433
审稿时长
1 months
期刊介绍: Theranostics serves as a pivotal platform for the exchange of clinical and scientific insights within the diagnostic and therapeutic molecular and nanomedicine community, along with allied professions engaged in integrating molecular imaging and therapy. As a multidisciplinary journal, Theranostics showcases innovative research articles spanning fields such as in vitro diagnostics and prognostics, in vivo molecular imaging, molecular therapeutics, image-guided therapy, biosensor technology, nanobiosensors, bioelectronics, system biology, translational medicine, point-of-care applications, and personalized medicine. Encouraging a broad spectrum of biomedical research with potential theranostic applications, the journal rigorously peer-reviews primary research, alongside publishing reviews, news, and commentary that aim to bridge the gap between the laboratory, clinic, and biotechnology industries.
×
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