4.8 Article

Neuroinvasive Listeria monocytogenes Infection Triggers IFN-Activation of Microglia and Upregulates Microglial miR-155

期刊

FRONTIERS IN IMMUNOLOGY
卷 9, 期 -, 页码 -

出版社

FRONTIERS MEDIA SA
DOI: 10.3389/fimmu.2018.02751

关键词

microRNA; meningitis; brain inflammation; microglia; interferon; Listeria

资金

  1. NIH from the National Institute of Neurological Diseases and Stroke [NS082639]
  2. Institutional Development Award (IDeA) from the National Institute of General Medical Sciences [GM103447]
  3. Presbyterian Health Foundation
  4. IDeA Program Infrastructure for Clinical and Translational Research from the National Institute of General Medical Sciences [1 U54GM104938]
  5. IDSA ERF Medical Scholars Program
  6. ASM Student Travel Grant
  7. Institutional Development Award (IDeA) from the National Institute of General Medical Sciences of the NIH [P20 GM103639]

向作者/读者索取更多资源

MicroRNA (miR) miR-155 modulates microglial activation and polarization, but its role in activation of microglia during bacterial brain infection is unclear. We studied miR-155 expression in brains of C57BL/6 (B6.WT) mice infected i.p. with the neuro-invasive bacterial pathogen Listeria monocytogenes (L. monocytogenes). Infected mice were treated with ampicillin starting 2 days (d) post-infection (p.i.) and analyzed 3d, 7d, and 14d p.i. Virulent L. monocytogenes strains EGD and 10403s upregulated miR-155 in whole brain 7 d p.i. whereas infection with avirulent, non-neurotropic Delta hly or Delta actA L. monocytogenes mutants did not. Similarly, infection with virulent but not mutated bacteria upregulated IFN-gamma mRNA in the brain at 7 d p.i. Upregulation of miR-155 in microglia was confirmed by qPCR of flow cytometry-sorted CD45(int)CD11b(pos) brain cells. Subsequently, brain leukocyte influxes and gene expression in sorted microglia were compared in L. monocytogenes-infected B6.WT and B6.Cg-Mir155tm1.1Rsky/J (B6.miR-155(-/-)) mice. Brain influxes of Ly-6C(high) monocytes and upregulation of IFN-related genes in microglia were similar to B6.WT mice at 3 d p.i. In contrast, by d 7 p.i. expressions of microglial IFN-related genes, including markers of M1 polarization, were significantly lower in B6.miR-155(-/-) mice and by 14 d p.i., influxes of activated T-lymphocytes were markedly reduced. Notably, CD45(high)CD11b(pos) brain cells from B6. miR-155(-/-)mice isolated at 7 d p.i. expressed 2-fold fewer IFN-gamma transcripts than did cells from B6. WT mice suggesting reduced IFN-gamma stimulation contributed to dampened gene expression in B6.miR-155(-/-) microglia. Lastly, in vitro stimulation of 7 d p.i. brain cells with heat-killed L. monocytogenes induced greater production of TNF in B6.miR-155(-/-) microglia than in B6.WT microglia. Thus, miR-155 affects brain inflammation by multiple mechanisms during neuroinvasive L. monocytogenes infection. Peripheral miR-155 promotes brain inflammation through its required role in optimal development of IFN-gamma-secreting lymphocytes that enter the brain and activate microglia. Microglial miR-155 promotes M1 polarization, and also inhibits inflammatory responses to stimulation by heat-killed L. monocytogenes, perhaps by targeting Tab2

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

4.8
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