Uracil DNA glycosylase counteracts APOBEC3G-induced hypermutation of hepatitis B viral genomes: excision repair of covalently closed circular DNA

PLoS Pathog. 2013;9(5):e1003361. doi: 10.1371/journal.ppat.1003361. Epub 2013 May 16.

Abstract

The covalently closed circular DNA (cccDNA) of the hepatitis B virus (HBV) plays an essential role in chronic hepatitis. The cellular repair system is proposed to convert cytoplasmic nucleocapsid (NC) DNA (partially double-stranded DNA) into cccDNA in the nucleus. Recently, antiviral cytidine deaminases, AID/APOBEC proteins, were shown to generate uracil residues in the NC-DNA through deamination, resulting in cytidine-to-uracil (C-to-U) hypermutation of the viral genome. We investigated whether uracil residues in hepadnavirus DNA were excised by uracil-DNA glycosylase (UNG), a host factor for base excision repair (BER). When UNG activity was inhibited by the expression of the UNG inhibitory protein (UGI), hypermutation of NC-DNA induced by either APOBEC3G or interferon treatment was enhanced in a human hepatocyte cell line. To assess the effect of UNG on the cccDNA viral intermediate, we used the duck HBV (DHBV) replication model. Sequence analyses of DHBV DNAs showed that cccDNA accumulated G-to-A or C-to-T mutations in APOBEC3G-expressing cells, and this was extensively enhanced by UNG inhibition. The cccDNA hypermutation generated many premature stop codons in the P gene. UNG inhibition also enhanced the APOBEC3G-mediated suppression of viral replication, including reduction of NC-DNA, pre-C mRNA, and secreted viral particle-associated DNA in prolonged culture. Enhancement of APOBEC3G-mediated suppression by UNG inhibition was not observed when the catalytic site of APOBEC3G was mutated. Transfection experiments of recloned cccDNAs revealed that the combination of UNG inhibition and APOBEC3G expression reduced the replication ability of cccDNA. Taken together, these data indicate that UNG excises uracil residues from the viral genome during or after cccDNA formation in the nucleus and imply that BER pathway activities decrease the antiviral effect of APOBEC3-mediated hypermutation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • APOBEC-3G Deaminase
  • Cytidine Deaminase / genetics
  • Cytidine Deaminase / metabolism*
  • DNA Repair*
  • DNA, Circular / genetics
  • DNA, Circular / metabolism*
  • DNA, Viral / genetics
  • DNA, Viral / metabolism*
  • Genome, Viral*
  • Hep G2 Cells
  • Hepatitis B virus / genetics
  • Hepatitis B virus / metabolism*
  • Humans
  • Mutation*
  • Uracil-DNA Glycosidase / genetics
  • Uracil-DNA Glycosidase / metabolism*

Substances

  • DNA, Circular
  • DNA, Viral
  • Uracil-DNA Glycosidase
  • APOBEC-3G Deaminase
  • APOBEC3G protein, human
  • Cytidine Deaminase

Grants and funding

This work was supported by the Funding Program for Next Generation World-Leading Researchers (NEXT Program) Grant Number LS051 (to MM), and JSPS KAKENHI Grant Number 21790654, 23790780 (to KK). The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.