Elsevier

Antiviral Research

Volume 149, January 2018, Pages 191-201
Antiviral Research

HBsAg mRNA degradation induced by a dihydroquinolizinone compound depends on the HBV posttranscriptional regulatory element

https://doi.org/10.1016/j.antiviral.2017.11.009Get rights and content

Highlights

  • A small molecule, DHQ-1, inhibited HBV surface protein production and viral replication.

  • DHQ-1 increased HBV surface mRNA turnover in the nucleus.

  • DHQ-1 induced viral mRNA turnover was dependent on the HBV Post-Transcriptional Regulatory Element (HPRE).

Abstract

In pursuit of novel therapeutics targeting the hepatitis B virus (HBV) infection, we evaluated a dihydroquinolizinone compound (DHQ-1) that in the nanomolar range reduced the production of virion and surface protein (HBsAg) in tissue culture. This compound also showed broad HBV genotype coverage, but was inactive against a panel of DNA and RNA viruses of other species. Oral administration of DHQ-1 in the AAV-HBV mouse model resulted in a significant reduction of serum HBsAg as soon as 4 days following the commencement of treatment. Reduction of HBV markers in both in vitro and in vivo experiments was related to the reduced amount of viral RNA including pre-genomic RNA (pgRNA) and 2.4/2.1 kb HBsAg mRNA. Nuclear run-on and subcellular fractionation experiments indicated that DHQ-1 mediated HBV RNA reduction was the result of accelerated viral RNA degradation in the nucleus, rather than the consequence of inhibition of transcription initiation. Through mutagenesis of HBsAg gene sequences, we found induction of HBsAg mRNA decay by DHQ-1 required the presence of the HBV posttranscriptional regulatory element (HPRE), with a 109 nucleotides sequence within the central region of the HPRE alpha sub-element being the most critical. Taken together, the current study shows that a small molecule can reduce the overall levels of HBV RNA, especially the HBsAg mRNA, and viral surface proteins. This may shed light on the development of a new class of HBV therapeutics.

Introduction

Chronic hepatitis B virus (HBV) infection affects more than 250 million people worldwide and is the major etiology of severe liver diseases such as fulminant hepatitis, hepatic cirrhosis and liver cancer (Liang et al., 2015). Although interferon alpha (IFNα) and nucleos(t)ide analogues (NA) are approved for the management of chronic HBV infection, a cure for the disease remains difficult to achieve (Lok et al., 2016, Terrault et al., 2016). Regarding IFNα treatment, sustained virologic response is only seen in 30%–40% selected patients, and is quiet often associated with severe side effects (Janssen et al., 2005, Perrillo, 2009). On the other hand, NAs are potent in the inhibition of HBV replication and are generally well tolerated. However, treatment with NAs may last lifelong and development of drug resistance could occur. Both IFNα and NAs are known for their low efficiency to induce hepatitis B virus surface antigen (HBsAg) seroconversion even after long-term treatment. In the blood, HBsAg exists as small lipid vesicles with an approximate size of 22 nm, and outnumbers virion particles 1000 to 100,000 times. The role of HBsAg in the maintenance of HBV infection is not clear. It is speculated that HBsAg may suppress immune reactions against virus or virus infected cells. High level of HBsAg is thought to be responsible for T cell exhaustion and depletion (Bertoletti and Ferrari, 2012, Boni et al., 2007). Reduction of HBsAg in blood quite often predicts better prognosis for patients receiving IFNα treatment (Cornberg et al., 2017). Ideally, the disappearance of HBsAg followed by the emergence of anti-HBsAg antibodies would result in a sustained virological response to HBV, which is regarded as a sign of a functional cure (Hoofnagle et al., 2007, McMahon et al., 2016). Therefore, down regulation of HBsAg, which is both an essential viral protein and an immune suppressor, is thought to be an important therapeutic target (Chen et al., 2012).

HBsAg is composed of large, middle and small surface proteins. The large (L) surface protein is translated from the 2.4 kb PreS1 transcript. The middle (M) and small (S) surface proteins are translated from the 2.1 kb PreS2/S transcript. The 2.4 and 2.1 kb mRNAs share in frame the S ORF and the remaining 3′ UTR (Seeger and Mason, 2000). HBsAg mRNA is intronless but contains cryptic splicing donor and acceptor sites (Hass et al., 2005). In mammalian cells, the regulation of RNA export is tightly linked with splicing processes and RNA maturation (Wagner and Lykke-Andersen, 2002). In order to avoid the disruption of HBsAg synthesis due to aberrant RNA splicing HBV has managed to guarantee its intronless RNA to be transported out of the nucleus. Different to HIV, whose spliced RNA transportation depends on the interaction of Rev and RRE RNA sequence, HBV uses a cis-element at the 3′ region of the transcript called HBV posttranscriptional regulatory element (HPRE) for RNA transportation (Huang and Liang, 1993, Huang and Yen, 1994, Visootsat et al., 2015). Without the help of HPRE, HBsAg mRNA is found to be degraded in the nucleus. Moreover, HPRE mediated intronless RNA nuclear export does not require the involvement of any viral protein (Huang and Yen, 1995). HBV PRE contains approximately 450 nucleotides encompassing nt 1151 to 1582 and is divided into 3 sub-elements HPREα (nt 1151–1346), HPREβ1 (nt 1347–1457) and HPREβ2 (nt 1458–1582) (Schwalbe et al., 2008, Smith et al., 1998). Each sub-element is able to independently but partially support mRNA transportation. The full export capacity needs the three sub-elements to work synergistically to transport HBsAg mRNA to the cytoplasm (Schwalbe et al., 2008). There are not many host proteins that have been found to interact with HPRE. La protein is found to bind the HPREα region and plays a role to maintain pgRNA and HBsAg mRNA stability (Heise et al., 1999b). In addition, polypyrimidine tract binding protein (PTB1) is reported to be associated with HPREβ2 region and is important to HBsAg mRNA nuclear export (Zang et al., 2001). Peculiarly, although GAPDH is regarded as a cytoplasmic protein it is found to be bound with HPREβ2 sub-element in the nucleus with an unknown function (Zang et al., 1998). Recent bioinformatics analysis of 6495 mammalian hepadnaviruses has revealed that the HPRE sequence contains two conserved stem-loops: SLα and SLβ, located in HPREα and HPREβ1 sub-elements, respectively. Homologs of these two conserved sequences have not been found in remotely related hepadnaviruses such as avian hepatitis B viruses, or in human mRNA (Lim and Brown, 2016). It is therefore of great interest to explore whether these special HBV PRE structures can be targeted by small molecule compounds (Chen et al., 2014).

In the current study, we report that a small molecule dihydroquinolizinone, 6-R2-10-methoxy-9-R1-2-oxo-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]-isoquinoline-3-carboxylicacid (DHQ-1), is able to induce HBV RNA degradation in tissue culture and mouse models. The molecule does not affect replication of a range of viruses including HCV, RSV, HIV, CMV, HSV, and HRV and thus is not believed to activate the innate antiviral response pathway through which HBV RNA could be affected. Degradation of HBV pgRNA and HBsAg mRNA occurs in the hepatocyte nucleus and requires de novo synthesis of host proteins. Mutagenesis analysis of the HBsAg expressing vector revealed that a 109-nucleotide region of the HBV PRE alpha sub-element, which contains the conserved La binding site and the stem-loop SLα is required for DHQ-1 antiviral activity.

Section snippets

Small molecules

6-R2-10-methoxy-9-R1-2-oxo-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]-isoquinoline-3-carboxylic acid is referred to as only DHQ-1. Its enantiomer is DHQ-2 (Fig. 1A). DHQ-1 and DHQ-2 were both provided by Arbutus Biopharma and were referred to in patent application WO 2015/113990A1 (Han et al., 2015).

Cells, HBV transfections and infections

The HBV producing cell line HepG2.2.15 (Sells et al., 1987) was cultured in DMEM/F12 containing 10% fetal bovine serum (FBS) and 100 U/ml penicillin and 100 μg/ml streptomycin (Invitrogen). HepG2,

The dihydroquinolizinone (DHQ-1) molecule specifically inhibited HBV replication in tissue culture

DHQ-1 was first evaluated for its ability to reduce HBV related products, including virion, HBsAg and e antigen (HBeAg) in the supernatants of HepG2.2.15 cells. Confluent HepG2.2.15 cells were incubated with varying concentrations of DHQ-1 for 5 days, after which the levels of viral particle, HBsAg and HBeAg in the culture media were determined. As shown in Fig. 1B, dose-dependent inhibition of three viral markers were determined with EC50 of 0.3 nM, 2 nM and 9 nM for viral particle, HBsAg and

Discussion

A small dihydroquinolizinone molecule, called DHQ-1, appeared to cause repression of HBsAg in the culture medium, as well as viral replicative intermediate DNA produced from integrated genomes (Han et al., 2015). We further found that DHQ-1 acted at the level of RNA by accelerating the nuclear degradation of viral transcripts, and confirmed that the compound also reduced liver HBV RNA and serum HBsAg in an HBV mouse model when administered orally.

DHQ-1 mediated reduction of HBV transcripts

Acknowledgements

We thank Dr. Shuping Tong for providing HBV plasmids of genotypes A, B and C, Dr. Koichi Watashi for the HepG2-NTCPc4 cell line. This work was supported by grants from the Commonwealth of Pennsylvania, the Hepatitis B Foundation and Arbutus Biopharma.

References (54)

  • W.Q. Zang et al.

    Identification of glyceraldehyde-3-phosphate dehydrogenase as a cellular protein that binds to the hepatitis B virus posttranscriptional regulatory element

    Virology

    (1998)
  • W.Q. Zang et al.

    Distinct export pathway utilized by the hepatitis B virus posttranscriptional regulatory element

    Virology

    (1999)
  • H. Zhao et al.

    Inhibition of human parainfluenza virus-3 replication by interferon and human MxA

    Virology

    (1996)
  • A. Bertoletti et al.

    Innate and adaptive immune responses in chronic hepatitis B virus infections: towards restoration of immune control of viral infection

    Gut

    (2012)
  • C. Boni et al.

    Characterization of hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection

    J. Virol.

    (2007)
  • A. Chen et al.

    Prospects for inhibiting the post-transcriptional regulation of gene expression in hepatitis B virus

    World J. Gastroenterol.

    (2014)
  • B. Chi et al.

    A Sub-Element in PRE enhances nuclear export of intronless mRNAs by recruiting the TREX complex via ZC3H18

    Nucleic Acids Res.

    (2014)
  • S. Dion et al.

    Adeno-associated virus-mediated gene transfer leads to persistent hepatitis B virus replication in mice expressing HLA-A2 and HLA-DR1 molecules

    J. Virol.

    (2013)
  • J.E. Donello et al.

    The hepatitis B virus posttranscriptional regulatory element is composed of two subelements

    J. Virol.

    (1996)
  • E. Gordien et al.

    Inhibition of hepatitis B virus replication by the interferon-inducible MxA protein

    J. Virol.

    (2001)
  • F. Guo et al.

    STING agonists induce an innate antiviral immune response against hepatitis B virus

    Antimicrob. Agents Chemother.

    (2015)
  • H. Guo et al.

    Characterization of the intracellular deproteinized relaxed circular DNA of hepatitis B virus: an intermediate of covalently closed circular DNA formation

    J. Virol.

    (2007)
  • Han, X.J., Hassan; Jiang, Min; Liang, Chungen; Wang, Jianping; Wang, Yongguang; Wang, Zhangguo; Weikert, Robert James;...
  • M. Hass et al.

    Functional analysis of hepatitis B virus reactivating in hepatitis B surface antigen-negative individuals

    Hepatology

    (2005)
  • T. Heise et al.

    Hepatitis B virus RNA-binding proteins associated with cytokine-induced clearance of viral RNA from the liver of transgenic mice

    J. Virol.

    (1999)
  • T. Heise et al.

    La autoantigen specifically recognizes a predicted stem-loop in hepatitis B virus RNA

    J. Virol.

    (1999)
  • T. Heise et al.

    Characterization of nuclear RNases that cleave hepatitis B virus RNA near the La protein binding site

    J. Virol.

    (2001)
  • Cited by (41)

    • Targeting the hepatitis B cccDNA with a sequence-specific ARCUS nuclease to eliminate hepatitis B virus in vivo

      2022, Molecular Therapy
      Citation Excerpt :

      However, given the overlapping transcript arrangement of the HBV genome, the nuclease target site falls within the 3′ untranslated region (UTR) of the HBsAg transcript, which is upstream of the polyadenylation sequence that is shared across all HBV transcripts. Previous studies have indicated this region may be important for transcript stability and/or export of mRNA out of the nucleus.33–35 In fact, the region of the HBV genome where the ARCUS-POL site (bp 1,259–1,280) is located has been noted as hyperconserved across viral variants, suggesting a vital role for the virus.36,37

    • Host RNA quality control as a hepatitis B antiviral target

      2021, Antiviral Research
      Citation Excerpt :

      La binds to a predicted stem-loop structure at the 5′ end of the posttranscriptional regulatory element (PRE) of HBV RNA between nucleotides 1273 and 1290 that has been associated with nuclear to cytoplasmic transport of the viral transcripts (Heise et al., 1999, 2001). The PRE, especially the HSLα stem-loop structure between nt 1292–1321, is also critical for virus susceptibility to the DHQ compound, and for the association of PAPD5 and another zinc finger protein ZCCHC14 with HBV RNA (Zhou et al., 2018; Hyrina et al., 2019; Kim et al., 2020). As discussed, polyadenylation at the 3′ ends of cellular mRNA is common and believed to be associated with mRNA stability (Tudek et al., 2018; Simonsen and Levinson, 1983; Moolla et al., 2002).

    View all citing articles on Scopus
    View full text