Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: covidwho-1501541

ABSTRACT

Long disregarded as junk DNA or genomic dark matter, endogenous retroviruses (ERVs) have turned out to represent important components of the antiviral immune response. These remnants of once-infectious retroviruses not only regulate cellular immune activation, but may even directly target invading viral pathogens. In this Gem, we summarize mechanisms by which retroviral fossils protect us from viral infections. One focus will be on recent advances in the role of ERVs as regulators of antiviral gene expression.


Subject(s)
Endogenous Retroviruses/physiology , Retroelements , Virus Diseases/immunology , Animals , Endogenous Retroviruses/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Immunity, Cellular , Promoter Regions, Genetic , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Pattern Recognition/metabolism , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Viral Proteins/metabolism , Virion/metabolism , Virus Diseases/genetics , Virus Diseases/virology
2.
Cell Rep ; 35(7): 109126, 2021 05 18.
Article in English | MEDLINE | ID: covidwho-1222854

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evades most innate immune responses but may still be vulnerable to some. Here, we systematically analyze the impact of SARS-CoV-2 proteins on interferon (IFN) responses and autophagy. We show that SARS-CoV-2 proteins synergize to counteract anti-viral immune responses. For example, Nsp14 targets the type I IFN receptor for lysosomal degradation, ORF3a prevents fusion of autophagosomes and lysosomes, and ORF7a interferes with autophagosome acidification. Most activities are evolutionarily conserved. However, SARS-CoV-2 Nsp15 antagonizes IFN signaling less efficiently than the orthologs of closely related RaTG13-CoV and SARS-CoV-1. Overall, SARS-CoV-2 proteins counteract autophagy and type I IFN more efficiently than type II or III IFN signaling, and infection experiments confirm potent inhibition by IFN-γ and -λ1. Our results define the repertoire and selected mechanisms of SARS-CoV-2 innate immune antagonists but also reveal vulnerability to type II and III IFN that may help to develop safe and effective anti-viral approaches.


Subject(s)
COVID-19/virology , SARS-CoV-2/immunology , Viral Proteins/immunology , Animals , Antiviral Agents/pharmacology , Autophagosomes/immunology , Autophagy/immunology , COVID-19/immunology , Cell Line , Chlorocebus aethiops , Exoribonucleases/immunology , HEK293 Cells , HeLa Cells , Humans , Immune Evasion , Immunity, Innate , Interferon Type I/metabolism , Interferons/metabolism , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , SARS-CoV-2/pathogenicity , Vero Cells , Viral Nonstructural Proteins/immunology
3.
Nucleic Acids Res ; 48(19): 10890-10908, 2020 11 04.
Article in English | MEDLINE | ID: covidwho-817440

ABSTRACT

Although endogenous retroviruses (ERVs) are known to harbor cis-regulatory elements, their role in modulating cellular immune responses remains poorly understood. Using an RNA-seq approach, we show that several members of the ERV9 lineage, particularly LTR12C elements, are activated upon HIV-1 infection of primary CD4+ T cells. Intriguingly, HIV-1-induced ERVs harboring transcription start sites are primarily found in the vicinity of immunity genes. For example, HIV-1 infection activates LTR12C elements upstream of the interferon-inducible genes GBP2 and GBP5 that encode for broad-spectrum antiviral factors. Reporter assays demonstrated that these LTR12C elements drive gene expression in primary CD4+ T cells. In line with this, HIV-1 infection triggered the expression of a unique GBP2 transcript variant by activating a cryptic transcription start site within LTR12C. Furthermore, stimulation with HIV-1-induced cytokines increased GBP2 and GBP5 expression in human cells, but not in macaque cells that naturally lack the GBP5 gene and the LTR12C element upstream of GBP2. Finally, our findings suggest that GBP2 and GBP5 have already been active against ancient viral pathogens as they suppress the maturation of the extinct retrovirus HERV-K (HML-2). In summary, our findings uncover how human cells can exploit remnants of once-infectious retroviruses to regulate antiviral gene expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endogenous Retroviruses/genetics , Gene Expression Regulation/immunology , HIV Infections/genetics , Promoter Regions, Genetic , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , HEK293 Cells , HIV Infections/immunology , HIV-1 , Humans , Macaca mulatta , T-Lymphocyte Subsets/cytology
SELECTION OF CITATIONS
SEARCH DETAIL