Your browser doesn't support javascript.
loading
RNA m6 A modification enzymes shape innate responses to DNA by regulating interferon ß.
Rubio, Rosa M; Depledge, Daniel P; Bianco, Christopher; Thompson, Letitia; Mohr, Ian.
Afiliação
  • Rubio RM; Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
  • Depledge DP; Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
  • Bianco C; Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
  • Thompson L; Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
  • Mohr I; Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
Genes Dev ; 32(23-24): 1472-1484, 2018 12 01.
Article em En | MEDLINE | ID: mdl-30463905
ABSTRACT
Modification of mRNA by N6-adenosine methylation (m6A) on internal bases influences gene expression in eukaryotes. How the dynamic genome-wide landscape of m6A-modified mRNAs impacts virus infection and host immune responses remains poorly understood. Here, we show that type I interferon (IFN) production triggered by dsDNA or human cytomegalovirus (HCMV) is controlled by the cellular m6A methyltrasferase subunit METTL14 and ALKBH5 demethylase. While METTL14 depletion reduced virus reproduction and stimulated dsDNA- or HCMV-induced IFNB1 mRNA accumulation, ALKBH5 depletion had the opposite effect. Depleting METTL14 increased both nascent IFNB1 mRNA production and stability in response to dsDNA. In contrast, ALKBH5 depletion reduced nascent IFNB1 mRNA production without detectably influencing IFN1B mRNA decay. Genome-wide transcriptome profiling following ALKBH5 depletion identified differentially expressed genes regulating antiviral immune responses, while METTL14 depletion altered pathways impacting metabolic reprogramming, stress responses, and aging. Finally, we determined that IFNB1 mRNA was m6A-modified within both the coding sequence and the 3' untranslated region (UTR). This establishes that the host m6A modification machinery controls IFNß production triggered by HCMV or dsDNA. Moreover, it demonstrates that responses to nonmicrobial dsDNA in uninfected cells, which shape host immunity and contribute to autoimmune disease, are regulated by enzymes controlling m6A epitranscriptomic changes.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: DNA / Regulação da Expressão Gênica / Interferon beta / Sistema Imunitário / Imunidade Inata / Metiltransferases Limite: Animals / Humans Idioma: En Revista: Genes Dev Assunto da revista: BIOLOGIA MOLECULAR Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: DNA / Regulação da Expressão Gênica / Interferon beta / Sistema Imunitário / Imunidade Inata / Metiltransferases Limite: Animals / Humans Idioma: En Revista: Genes Dev Assunto da revista: BIOLOGIA MOLECULAR Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos