Your browser doesn't support javascript.
loading
Protein S-Nitrosylation of Human Cytomegalovirus pp71 Inhibits Its Ability To Limit STING Antiviral Responses.
Nukui, Masatoshi; Roche, Kathryn L; Jia, Jie; Fox, Paul L; Murphy, Eain A.
Affiliation
  • Nukui M; Microbiology and Immunology Department, SUNY Upstate Medical University, Syracuse, New York, USA.
  • Roche KL; Evrys Bio, Pennsylvania Biotechnology Center, Doylestown, Pennsylvania, USA.
  • Jia J; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.
  • Fox PL; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.
  • Murphy EA; Microbiology and Immunology Department, SUNY Upstate Medical University, Syracuse, New York, USA murphye1@upstate.edu.
J Virol ; 94(17)2020 08 17.
Article de En | MEDLINE | ID: mdl-32581105
ABSTRACT
Human Cytomegalovirus (HCMV) is a ubiquitous pathogen that has coevolved with its host and, in doing so, is highly efficient in undermining antiviral responses that limit successful infections. As a result, HCMV infections are highly problematic in individuals with weakened or underdeveloped immune systems, including transplant recipients and newborns. Understanding how HCMV controls the microenvironment of an infected cell so as to favor productive replication is of critical importance. To this end, we took an unbiased proteomics approach to identify the highly reversible, stress-induced, posttranslational modification (PTM) protein S-nitrosylation on viral proteins to determine the biological impact on viral replication. We identified protein S-nitrosylation of 13 viral proteins during infection of highly permissive fibroblasts. One of these proteins, pp71, is critical for efficient viral replication, as it undermines host antiviral responses, including stimulator of interferon genes (STING) activation. By exploiting site-directed mutagenesis of the specific amino acids we identified in pp71 as protein S-nitrosylated, we found this pp71 PTM diminishes its ability to undermine antiviral responses induced by the STING pathway. Our results suggest a model in which protein S-nitrosylation may function as a host response to viral infection that limits viral spread.IMPORTANCE In order for a pathogen to establish a successful infection, it must undermine the host cell responses inhibitory to the pathogen. As such, herpesviruses encode multiple viral proteins that antagonize each host antiviral response, thereby allowing for efficient viral replication. Human Cytomegalovirus encodes several factors that limit host countermeasures to infection, including pp71. Herein, we identified a previously unreported posttranslational modification of pp71, protein S-nitrosylation. Using site-directed mutagenesis, we mutated the specific sites of this modification thereby blocking this pp71 posttranslational modification. In contexts where pp71 is not protein S-nitrosylated, host antiviral response was inhibited. The net result of this posttranslational modification is to render a viral protein with diminished abilities to block host responses to infection. This novel work supports a model in which protein S-nitrosylation may be an additional mechanism in which a cell inhibits a pathogen during the course of infection.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Antiviraux / Protéines virales / Protéine S / Cytomegalovirus Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: J Virol Année: 2020 Type de document: Article Pays d'affiliation: États-Unis d'Amérique

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Antiviraux / Protéines virales / Protéine S / Cytomegalovirus Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: J Virol Année: 2020 Type de document: Article Pays d'affiliation: États-Unis d'Amérique