4.7 Article

The intestinal microbial metabolite nicotinamide n-oxide prevents herpes simplex encephalitis via activating mitophagy in microglia

期刊

GUT MICROBES
卷 14, 期 1, 页码 -

出版社

TAYLOR & FRANCIS INC
DOI: 10.1080/19490976.2022.2096989

关键词

HSE; gut microbiota; microglia; nicotinamide n-oxide; mitophagy

资金

  1. National Natural Science Foundation of China [81573471, 81872908, 82104238]
  2. Key Laboratory of Virology of Guangzhou [201705030003]
  3. Guangdong Basic and Applied Basic Research Foundation [2021B1515120088, 2020A151511056601]
  4. Guangzhou Major program of the Industry-University-Research collaborative innovation [201704030087, 201604020178]

向作者/读者索取更多资源

This study found that HSE mice exhibited dysbiosis of gut microbiota and altered metabolite configuration. The activation of microglia played an important role in HSE. Depletion of gut microbiota by antibiotic treatment triggered hyper-activation of microglia, leading to increased inflammatory immune response and viral burden. Exogenous administration of NAMO, a microbial metabolite, reduced microglia-mediated proinflammatory response and limited HSV-1 infection. NAMO was mainly generated by neomycin-sensitive bacteria.
Herpes simplex encephalitis (HSE), a complication of herpes simplex virus type I (HSV-1) infection causes neurological disorder or even death in immunocompromised adults and newborns. However, the intrinsic factors controlling the HSE outcome remain unclear. Here, we show that HSE mice exhibit gut microbiota dysbiosis and altered metabolite configuration and tryptophan-nicotinamide metabolism. HSV-1 neurotropic infection activated microglia, with changed immune properties and cell numbers, to stimulate antiviral immune response and contribute substantially to HSE. In addition, depletion of gut microbiota by oral antibiotics (ABX)-treatment triggered the hyper-activation of microglia, which in turn enhanced inflammatory immune response, and cytokine production, resulting in aggregated viral burden and HSE pathology. Furthermore, exogenous administration of nicotinamide n-oxide (NAMO), an oxidative product of nicotinamide derived from gut microbiota, to ABX-treated or untreated HSE mice significantly diminished microglia-mediated proinflammatory response and limited HSV-1 infection in CNS. Mechanistic study revealed that HSV-1 activates microglia by increasing mitochondrial damage via defective mitophagy, whereas microbial metabolite NAMO restores NAD+-dependent mitophagy to inhibit microglia activation and HSE progression. NAMO also prevented neuronal cell death triggered by HSV-1 infection or microglia-mediated microenvironmental toxicity. Finally, we show that NAMO is mainly generated by neomycin-sensitive bacteria, especially Lactobacillus_gasseri and Lactobacillus_reuteri. Together, these data demonstrate that gut microbial metabolites act as intrinsic restrictive factors against HSE progression via regulating mitophagy in microglia, implying further exploration of bacterial or nutritional approaches for treating neurotropic virus-related neurodegenerative diseases.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据