Zinc Finger Protein ZFP36L1 Inhibits Flavivirus Infection by both 5'-3' XRN1 and 3'-5' RNA-Exosome RNA Decay Pathways

J Virol. 2022 Jan 12;96(1):e0166521. doi: 10.1128/JVI.01665-21. Epub 2021 Oct 13.

Abstract

Zinc-finger protein 36, CCCH type-like 1 (ZFP36L1), containing tandem CCCH-type zinc-finger motifs with an RNA-binding property, plays an important role in cellular RNA metabolism mainly by RNA decay pathways. Recently, we demonstrated that human ZFP36L1 has potent antiviral activity against influenza A virus infection. However, its role in the host defense response against flaviviruses has not been addressed. Here, we demonstrate that ZFP36L1 functions as a host innate defender against flaviviruses, including Japanese encephalitis virus (JEV) and dengue virus (DENV). Overexpression of ZFP36L1 reduced JEV and DENV infection, and ZFP36L1 knockdown enhanced viral replication. ZFP36L1 destabilized the JEV genome by targeting and degrading viral RNA mediated by both 5'-3' XRN1 and 3'-5' RNA-exosome RNA decay pathways. Mutation in both zinc-finger motifs of ZFP36L1 disrupted RNA-binding and antiviral activity. Furthermore, the viral RNA sequences specifically recognized by ZFP36L1 were mapped to the 3'-untranslated region of the JEV genome with the AU-rich element (AUUUA) motif. We extend the function of ZFP36L1 to host antiviral defense by directly binding and destabilizing the viral genome via recruiting cellular mRNA decay machineries. IMPORTANCE Cellular RNA-binding proteins are among the first lines of defense against various viruses, particularly RNA viruses. ZFP36L1 belongs to the CCCH-type zinc-finger protein family and has RNA-binding activity; it has been reported to bind directly to the AU-rich elements (AREs) of a subset of cellular mRNAs and then lead to mRNA decay by recruiting mRNA-degrading enzymes. However, the antiviral potential of ZFP36L1 against flaviviruses has not yet been fully demonstrated. Here, we reveal the antiviral potential of human ZFP36L1 against Japanese encephalitis virus (JEV) and dengue virus (DENV). ZFP36L1 specifically targeted the ARE motif within viral RNA and triggered the degradation of viral RNA transcripts via cellular degrading enzymes 5'-3' XRN1 and 3'-5' RNA exosome. These findings provide mechanistic insights into how human ZFP36L1 serves as a host antiviral factor to restrict flavivirus replication.

Keywords: ZFP36L1; antiviral mechanism; flavivirus; zinc-finger RNA-binding protein.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • 3' Untranslated Regions
  • Amino Acid Motifs
  • Butyrate Response Factor 1 / chemistry
  • Butyrate Response Factor 1 / metabolism*
  • Dengue Virus / physiology
  • Encephalitis Virus, Japanese / physiology
  • Exoribonucleases / metabolism*
  • Exosome Multienzyme Ribonuclease Complex / metabolism*
  • Flavivirus / physiology*
  • Flavivirus Infections / metabolism*
  • Flavivirus Infections / virology*
  • Host-Pathogen Interactions
  • Humans
  • Microtubule-Associated Proteins / metabolism*
  • Protein Binding
  • Protein Interaction Domains and Motifs
  • RNA Stability*
  • RNA, Viral / genetics
  • RNA, Viral / metabolism
  • RNA-Binding Proteins
  • Virus Replication*

Substances

  • 3' Untranslated Regions
  • Butyrate Response Factor 1
  • Microtubule-Associated Proteins
  • RNA, Viral
  • RNA-Binding Proteins
  • ZFP36L1 protein, human
  • Exoribonucleases
  • Exosome Multienzyme Ribonuclease Complex
  • XRN1 protein, human