Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
1.
Immunity ; 55(11): 2103-2117.e10, 2022 Nov 08.
Article in English | MEDLINE | ID: covidwho-2095502

ABSTRACT

The surface of the central nervous system (CNS) is protected by the meninges, which contain a dense network of meningeal macrophages (MMs). Here, we examined the role of tissue-resident MM in viral infection. MHC-II- MM were abundant neonatally, whereas MHC-II+ MM appeared over time. These barrier macrophages differentially responded to in vivo peripheral challenges such as LPS, SARS-CoV-2, and lymphocytic choriomeningitis virus (LCMV). Peripheral LCMV infection, which was asymptomatic, led to a transient infection and activation of the meninges. Mice lacking macrophages but conserving brain microglia, or mice bearing macrophage-specific deletion of Stat1 or Ifnar, exhibited extensive viral spread into the CNS. Transcranial pharmacological depletion strategies targeting MM locally resulted in several areas of the meninges becoming infected and fatal meningitis. Low numbers of MHC-II+ MM, which is seen upon LPS challenge or in neonates, corelated with higher viral load upon infection. Thus, MMs protect against viral infection and may present targets for therapeutic manipulation.


Subject(s)
COVID-19 , Lymphocytic Choriomeningitis , Animals , Mice , Lipopolysaccharides , Mice, Inbred C57BL , SARS-CoV-2 , Lymphocytic choriomeningitis virus/physiology , Macrophages , Meninges
2.
Sci Adv ; 8(6): eabk2691, 2022 02 11.
Article in English | MEDLINE | ID: covidwho-1685473

ABSTRACT

Upon virus infection, CD8+ T cell accumulation is tightly controlled by simultaneous proliferation and apoptosis. However, it remains unclear how TCR signal coordinates these events to achieve expansion and effector cell differentiation. We found that T cell-specific deletion of nuclear helicase Dhx9 led to impaired CD8+ T cell survival, effector differentiation, and viral clearance. Mechanistically, Dhx9 acts as the key regulator to ensure LCK- and CD3ε-mediated ZAP70 phosphorylation and ERK activation to protect CD8+ T cells from apoptosis before proliferative burst. Dhx9 directly regulates Id2 transcription to control effector CD8+ T cell differentiation. The DSRM and OB_Fold domains are required for LCK binding and Id2 transcription, respectively. Dhx9 expression is predominantly increased in effector CD8+ T cells of COVID-19 patients. Therefore, we revealed a previously unknown regulatory mechanism that Dhx9 protects activated CD8+ T cells from apoptosis and ensures effector differentiation to promote antiviral immunity independent of nuclear sensor function.


Subject(s)
Antiviral Agents/pharmacology , Arenaviridae Infections/prevention & control , CD8-Positive T-Lymphocytes/immunology , COVID-19/prevention & control , DEAD-box RNA Helicases/metabolism , Immunity, Innate , Neoplasm Proteins/metabolism , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , Arenaviridae Infections/pathology , COVID-19/immunology , COVID-19/metabolism , COVID-19/pathology , Cell Differentiation , DEAD-box RNA Helicases/genetics , Humans , Lymphocyte Activation , Lymphocytic choriomeningitis virus/physiology , Mice , Neoplasm Proteins/genetics , SARS-CoV-2/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL