关键词: IL-12 dendritic cells mtDNA myeloid STING

Mesh : Animals Dendritic Cells / immunology metabolism Membrane Proteins / metabolism genetics Mice Interleukin-12 / metabolism genetics DNA, Mitochondrial / genetics metabolism Mice, Knockout Intestinal Mucosa / metabolism pathology immunology Mice, Inbred C57BL Colitis / pathology chemically induced metabolism genetics Signal Transduction Colitis, Ulcerative / genetics pathology metabolism immunology Colitis-Associated Neoplasms / pathology genetics metabolism immunology Macrophages / metabolism immunology Disease Models, Animal Dextran Sulfate

来  源:   DOI:10.7150/thno.96184   PDF(Pubmed)

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.
摘要:
原理:溃疡性结肠炎(UC)的治疗提出了持续的临床挑战。新兴研究暗示cGAS-STING途径促进UC的进展,但是矛盾的结果阻碍了STING作为治疗靶标的发展。在目前的研究中,我们旨在全面阐明其起源,髓样STING在结肠炎和结肠炎相关癌(CAC)中的下游信号传导和致病作用。方法:构建Tmem173fl/flLyz2-Creert2小鼠,用于诱导骨髓特异性缺失STING。RNA测序,流式细胞术,和多重免疫组织化学用于研究DSS诱导的结肠炎或AOM/DSS诱导的癌变中的免疫反应。结肠类器官,原代骨髓源性巨噬细胞和树突状细胞,和脾T细胞用于体外研究。结果:我们观察到成年小鼠的髓样STING敲除抑制巨噬细胞成熟,减少DC细胞激活,并抑制促炎Th1和Th17细胞,从而防止急性和慢性结肠炎和CAC。然而,新生小鼠或肿瘤小鼠的髓样STING缺失表现出受损的免疫耐受和抗肿瘤免疫力。此外,我们发现从受损的结肠类器官释放的TFAM相关mtDNA,而不是细菌产品,以细胞外囊泡非依赖性但内吞依赖性方式激活树突状细胞中的STING。IRF3和NF-κB都是STING介导的IL-12家族细胞因子表达所必需的,促进Th1和Th17分化,并有助于结肠炎的过度炎症。结论:通过髓样STING检测受损肠上皮的TFAM-mtDNA复合物可通过IL-12细胞因子加重结肠炎,提供新的证据支持STING作为UC和CAC的治疗靶点的发展。
公众号