Your browser doesn't support javascript.
loading
Respiratory Syncytial Virus Matrix Protein-Chromatin Association Is Key to Transcriptional Inhibition in Infected Cells.
Li, Hong-Mei; Ghildyal, Reena; Hu, Mengjie; Tran, Kim C; Starrs, Lora M; Mills, John; Teng, Michael N; Jans, David A.
Affiliation
  • Li HM; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Melbourne, VIC 3800, Australia.
  • Ghildyal R; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Melbourne, VIC 3800, Australia.
  • Hu M; Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia.
  • Tran KC; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Melbourne, VIC 3800, Australia.
  • Starrs LM; Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
  • Mills J; Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia.
  • Teng MN; Department of Infectious Diseases, School of Biomedical Sciences, Monash University and the Burnet Institute, Melbourne, VIC 3004, Australia.
  • Jans DA; Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
Cells ; 10(10)2021 10 18.
Article in En | MEDLINE | ID: mdl-34685766
ABSTRACT
The morbidity and mortality caused by the globally prevalent human respiratory pathogen respiratory syncytial virus (RSV) approaches that world-wide of influenza. We previously demonstrated that the RSV matrix (M) protein shuttles, in signal-dependent fashion, between host cell nucleus and cytoplasm, and that this trafficking is central to RSV replication and assembly. Here we analyze in detail the nuclear role of M for the first time using a range of novel approaches, including quantitative analysis of de novo cell transcription in situ in the presence or absence of RSV infection or M ectopic expression, as well as in situ DNA binding. We show that M, dependent on amino acids 110-183, inhibits host cell transcription in RSV-infected cells as well as cells transfected to express M, with a clear correlation between nuclear levels of M and the degree of transcriptional inhibition. Analysis of bacterially expressed M protein and derivatives thereof mutated in key residues within M's RNA binding domain indicates that M can bind to DNA as well as RNA in a cell-free system. Parallel results for point-mutated M derivatives implicate arginine 170 and lysine 172, in contrast to other basic residues such as lysine 121 and 130, as critically important residues for inhibition of transcription and DNA binding both in situ and in vitro. Importantly, recombinant RSV carrying arginine 170/lysine 172 mutations shows attenuated infectivity in cultured cells and in an animal model, concomitant with altered inflammatory responses. These findings define an RSV M-chromatin interface critical for host transcriptional inhibition in infection, with important implications for anti-RSV therapeutic development.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription, Genetic / Chromatin / Viral Matrix Proteins / Respiratory Syncytial Virus, Human / Respiratory Syncytial Virus Infections Type of study: Prognostic_studies / Risk_factors_studies Limits: Animals / Humans Language: En Journal: Cells Year: 2021 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription, Genetic / Chromatin / Viral Matrix Proteins / Respiratory Syncytial Virus, Human / Respiratory Syncytial Virus Infections Type of study: Prognostic_studies / Risk_factors_studies Limits: Animals / Humans Language: En Journal: Cells Year: 2021 Document type: Article Affiliation country: