Downregulation of microRNA miR-526a by enterovirus inhibits RIG-I-dependent innate immune response

J Virol. 2014 Oct;88(19):11356-68. doi: 10.1128/JVI.01400-14. Epub 2014 Jul 23.

Abstract

Retinoic acid-inducible gene I (RIG-I) is an intracellular RNA virus sensor that induces type I interferon-mediated host-protective innate immunity against viral infection. Although cylindromatosis (CYLD) has been shown to negatively regulate innate antiviral response by removing K-63-linked polyubiquitin from RIG-I, the regulation of its expression and the underlying regulatory mechanisms are still incompletely understood. Here we show that RIG-I activity is regulated by inhibition of CYLD expression mediated by the microRNA miR-526a. We found that viral infection specifically upregulates miR-526a expression in macrophages via interferon regulatory factor (IRF)-dependent mechanisms. In turn, miR-526a positively regulates virus-triggered type I interferon (IFN-I) production, thus suppressing viral replication, the underlying mechanism of which is the enhancement of RIG-I K63-linked ubiquitination by miR-526a via suppression of the expression of CYLD. Remarkably, virus-induced miR-526a upregulation and CYLD downregulation are blocked by enterovirus 71 (EV71) 3C protein, while ectopic miR-526a expression inhibits the replication of EV71 virus. The collective results of this study suggest a novel mechanism of the regulation of RIG-I activity during RNA virus infection by miR-526a and suggest a novel mechanism for the evasion of the innate immune response controlled by EV71.

Importance: RNA virus infection upregulates the expression of miR-526a in macrophages through IRF-dependent pathways. In turn, miR-526a positively regulates virus-triggered type I IFN production and inhibits viral replication, the underlying mechanism of which is the enhancement of RIG-I K-63 ubiquitination by miR-526a via suppression of the expression of CYLD. Remarkably, virus-induced miR-526a upregulation and CYLD downregulation are blocked by enterovirus 71 (EV71) 3C protein; cells with overexpressed miR-526a were highly resistant to EV71 infection. The collective results of this study suggest a novel mechanism of the regulation of RIG-I activity during RNA virus infection by miR-526a and propose a novel mechanism for the evasion of the innate immune response controlled by EV71.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • 3C Viral Proteases
  • Animals
  • Chlorocebus aethiops
  • Cysteine Endopeptidases / genetics
  • Cysteine Endopeptidases / immunology
  • DEAD Box Protein 58
  • DEAD-box RNA Helicases / genetics*
  • DEAD-box RNA Helicases / immunology
  • Deubiquitinating Enzyme CYLD
  • Dogs
  • Enterovirus A, Human / genetics*
  • Enterovirus A, Human / immunology
  • Gene Expression Regulation
  • HEK293 Cells
  • Host-Pathogen Interactions
  • Humans
  • Immune Evasion*
  • Immunity, Innate*
  • Interferon Type I / genetics
  • Interferon Type I / immunology
  • Macrophages / immunology
  • Macrophages / virology
  • Madin Darby Canine Kidney Cells
  • MicroRNAs / genetics*
  • MicroRNAs / immunology
  • Polyubiquitin / genetics
  • Polyubiquitin / immunology
  • Receptors, Immunologic
  • Signal Transduction
  • Tumor Suppressor Proteins / genetics
  • Tumor Suppressor Proteins / immunology
  • Vero Cells
  • Viral Proteins / genetics*
  • Viral Proteins / immunology
  • Virus Replication

Substances

  • Interferon Type I
  • MicroRNAs
  • Receptors, Immunologic
  • Tumor Suppressor Proteins
  • Viral Proteins
  • Polyubiquitin
  • CYLD protein, human
  • Deubiquitinating Enzyme CYLD
  • Cysteine Endopeptidases
  • 3C Viral Proteases
  • RIGI protein, human
  • DEAD Box Protein 58
  • DEAD-box RNA Helicases