Back to Search Start Over

Hepatitis B virus X protein counteracts high mobility group box 1 protein-mediated epigenetic silencing of covalently closed circular DNA.

Authors :
Kim, Elena S.
Zhou, Jun
Zhang, Hu
Marchetti, Alexander
van de Klundert, Maarten
Cai, Dawei
Yu, Xiaoyang
Mitra, Bidisha
Liu, Yuanjie
Wang, Mu
Protzer, Ulrike
Guo, Haitao
Source :
PLoS Pathogens. 6/9/2022, Vol. 18 Issue 6, p1-31. 31p.
Publication Year :
2022

Abstract

Hepatitis B virus (HBV) covalently closed circular DNA (cccDNA), serving as the viral persistence form and transcription template of HBV infection, hijacks host histone and non-histone proteins to form a minichromosome and utilizes posttranslational modifications (PTMs) "histone code" for its transcriptional regulation. HBV X protein (HBx) is known as a cccDNA transcription activator. In this study we established a dual system of the inducible reporter cell lines modelling infection with wildtype (wt) and HBx-null HBV, both secreting HA-tagged HBeAg as a semi-quantitative marker for cccDNA transcription. The cccDNA-bound histone PTM profiling of wt and HBx-null systems, using chromatin immunoprecipitation coupled with quantitative PCR (ChIP-qPCR), confirmed that HBx is essential for maintenance of cccDNA at transcriptionally active state, characterized by active histone PTM markers. Differential proteomics analysis of cccDNA minichromosome established in wt and HBx-null HBV cell lines revealed group-specific hits. One of the hits in HBx-deficient condition was a non-histone host DNA-binding protein high mobility group box 1 (HMGB1). Its elevated association to HBx-null cccDNA was validated by ChIP-qPCR assay in both the HBV stable cell lines and infection systems in vitro. Furthermore, experimental downregulation of HMGB1 in HBx-null HBV inducible and infection models resulted in transcriptional re-activation of the cccDNA minichromosome, accompanied by a switch of the cccDNA-associated histones to euchromatic state with activating histone PTMs landscape and subsequent upregulation of cccDNA transcription. Mechanistically, HBx interacts with HMGB1 and prevents its binding to cccDNA without affecting the steady state level of HMGB1. Taken together, our results suggest that HMGB1 is a novel host restriction factor of HBV cccDNA with epigenetic silencing mechanism, which can be counteracted by viral transcription activator HBx. Author summary: Nowadays hepatitis B virus (HBV) prevalence remains a significant burden to the worldwide healthcare. There is no cure for chronic HBV infection so far due to the lack of therapies that enable elimination of an intrahepatic persistent form of HBV DNA genome, namely, the covalently closed circular DNA (cccDNA). Inside the infected liver cells, cccDNA hijacks host structural and regulatory factors to assemble into a minichromosome, but certain intrinsic host factors are able to counteract cccDNA activity. In our study, we identified an anti-HBV host restriction factor, specifically the high mobility group box 1 protein (HMGB1), and demonstrated its potential to mediate an epigenetic silencing, i.e. functional inactivation, of the cccDNA. However, the virally encoded accessory protein HBx is able to antagonize HMGB1 and maintain an active state of cccDNA. This may contribute to a better understanding of virus-host interaction during HBV infection, and to the development of HBV infection epigenetic drugs and re-consideration of cancer therapeutics strategies, where HMGB1 is used as an anti-cancer target. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
15537366
Volume :
18
Issue :
6
Database :
Academic Search Index
Journal :
PLoS Pathogens
Publication Type :
Academic Journal
Accession number :
157359058
Full Text :
https://doi.org/10.1371/journal.ppat.1010576