当前位置: X-MOL 学术Cell. Mol. Immunol. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
DNA sensing and associated type 1 interferon signaling contributes to progression of radiation-induced liver injury
Cellular & Molecular Immunology ( IF 24.1 ) Pub Date : 2020-03-19 , DOI: 10.1038/s41423-020-0395-x
Shisuo Du 1 , Genwen Chen 1 , Baoying Yuan 1 , Yong Hu 1 , Ping Yang 1 , Yixing Chen 1 , Qianqian Zhao 1 , Jian Zhou 2 , Jia Fan 2 , Zhaochong Zeng 1
Affiliation  

Liver damage upon exposure to ionizing radiation (IR), whether accidental or therapeutic, can contribute to liver dysfunction. Currently, radiotherapy (RT) is used for various cancers including hepatocellular carcinoma (HCC); however, the treatment dose is limited by radiation-induced liver disease (RILD) with a high mortality rate. Furthermore, the precise molecular mechanisms of RILD remain poorly understood. Here, we investigated RILD pathogenesis using various knockout mouse strains subjected to whole-liver irradiation. We found that hepatocytes released a large quantity of double-stranded DNA (dsDNA) after irradiation. The cGAS-STING pathway in non-parenchymal cells (NPCs) was promptly activated by this dsDNA, causing interferon (IFN)-I production and release and concomitant hepatocyte damage. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against RILD. Moreover, clinically irradiated human peri-HCC liver tissues exhibited substantially higher STING and IFNβ expression than non-irradiated tissues. Increased serum IFNβ concentrations post-radiation were associated with RILD development in patients. These results delineate cGAS-STING induced type 1 interferon release in NPCs as a key mediator of IR-induced liver damage and described a mechanism of innate-immunity-driven pathology, linking cGAS-STING activation with amplification of initial radiation-induced liver injury.



中文翻译:

DNA 传感和相关的 1 型干扰素信号传导有助于放射性肝损伤的进展

暴露于电离辐射 (IR) 后的肝损伤,无论是意外的还是治疗性的,都可能导致肝功能障碍。目前,放疗(RT)被用于各种癌症,包括肝细胞癌(HCC);然而,治疗剂量受限于高死亡率的放射性肝病(RILD)。此外,RILD 的精确分子机制仍然知之甚少。在这里,我们使用各种经过全肝照射的敲除小鼠品系研究了 RILD 发病机制。我们发现肝细胞在照射后会释放出大量的双链 DNA(dsDNA)。非实质细胞 (NPC) 中的 cGAS-STING 通路被这种 dsDNA 迅速激活,导致干扰素 (IFN)-I 的产生和释放以及伴随的肝细胞损伤。IFN-I 信号通路的遗传和药理学消融保护免受 RILD。此外,临床辐照的人 HCC 周围肝组织表现出比未辐照组织显着更高的 STING 和 IFNβ 表达。放射后血清 IFNβ 浓度增加与患者的 RILD 发展相关。这些结果将 cGAS-STING 诱导的 NPC 中 1 型干扰素释放描述为 IR 诱导的肝损伤的关键介质,并描述了先天免疫驱动的病理学机制,将 cGAS-STING 激活与初始辐射诱导的肝损伤的放大联系起来。放射后血清 IFNβ 浓度增加与患者的 RILD 发展相关。这些结果将 cGAS-STING 诱导的 NPC 中 1 型干扰素释放描述为 IR 诱导的肝损伤的关键介质,并描述了先天免疫驱动的病理学机制,将 cGAS-STING 激活与初始辐射诱导的肝损伤的放大联系起来。放射后血清 IFNβ 浓度增加与患者的 RILD 发展相关。这些结果将 cGAS-STING 诱导的 NPC 中 1 型干扰素释放描述为 IR 诱导的肝损伤的关键介质,并描述了先天免疫驱动的病理学机制,将 cGAS-STING 激活与初始辐射诱导的肝损伤的放大联系起来。

更新日期:2020-04-24
down
wechat
bug