You are on page 1of 10

Antiviral Research 182 (2020) 104925

Contents lists available at ScienceDirect

Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral

Invited Review

Hepatitis B virus biology and life cycle


Senko Tsukuda a, b, Koichi Watashi a, c, d, e, *
a
Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
b
Nuffield Department of Medicine, University of Oxford, Oxford, UK
c
Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
d
Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
e
MIRAI, JST, Saitama, Japan

A R T I C L E I N F O A B S T R A C T

Keywords: Hepatitis B virus (HBV) specifically infects hepatocytes and causes severe liver diseases. The HBV life cycle is
HBV unique in that the genomic DNA (relaxed-circular partially double-stranded DNA: rcDNA) is converted to a
Life cycle molecular template DNA (covalently closed circular DNA: cccDNA) to amplify a viral RNA intermediate, which is
Entry
then reverse-transcribed back to viral DNA. The highly stable characteristics of cccDNA result in chronic
Replication
cccDNA
infection and a poor rate of cure. This complex life cycle of HBV offers a variety of targets to develop antiviral
NTCP agents. We provide here an update on the current knowledge of HBV biology and its life cycle, which may help to
identify new antiviral targets.

(naked nucleocapsids) (Hu and Liu, 2017).


The HBV genome DNA is a relaxed-circular DNA (rcDNA) of
1. Introduction approximately 3.2 kb in length with a complete minus strand and an
incomplete plus strand (Fig. 2). The viral genome encodes four over­
Hepatitis B virus (HBV) is an enveloped DNA virus, classified in the lapping open reading frames (ORFs), C, P, S, and X, from which func­
Hepadnaviridae (Rajoriya et al., 2017). HBV was first discovered in an tional viral proteins are produced: HBc and its relatives such as E antigen
Australian aborigine by the detection of its antigen, currently known as (HBe) and 22-kDa precore protein (p22cr) from C; Pol from P; three
surface antigen and originally called “Australia antigen” as reported by types of surface antigens, L-HBs, M-HBs, and S-HBs, from S; and HBV X
Blumberg and colleagues in 1965 (Blumberg et al., 1965). The presence protein (HBx) from X. rcDNA is converted into covalently closed circular
of this antigen in hepatitis patients was independently reported by DNA (cccDNA) in infected cells (see below), and cccDNA produces HBV
Prince and by Okochi and Murakami in 1968 (Okochi and Murakami, RNAs of different lengths (mainly 3.5 Kb, 2.4 Kb, 2.1 Kb, and 0.7 Kb)
1968; Prince, 1968), and the virus particles were visualized by an transcribed from different promoters in the HBV genome. A 3.5 Kb RNA
electron microscope in 1970 by Dane and colleagues (Dane et al., 1970). produces the protein product from C and P; a 2.4 Kb RNA is translated
At least three types of HBV particles are observed in the serum of into L-HBs and a 2.1 Kb RNA synthesizes the other two surface antigens,
infected patients: spherical structures of 42 nm in diameter, those with a M-HBs and S-HBs; and a 0.7 Kb RNA produces HBx. The produced HBc
diameter of 22 nm, and filament structures of variable length that are 22 protein product initially associates to yield a dimer through its N-ter­
nm in a diameter (Fig. 1). The 42 nm particles, also called Dane particles, minal domain, and then self-assembles into an icosahedral capsid con­
are infectious virions consisting of a lipid membrane with three viral sisting of 90 or 120 dimers, which incorporates the 3.5 Kb viral
surface antigens (HBs), large (L-HBs), middle (M-HBs), and small pregenomic RNA (pgRNA) associated with Pol (see below in detail). HBe
(S-HBs), that surround a nucleocapsid composed of hepatitis B core is produced by the translation of 3.5 Kb preC mRNA, having an extended
protein (HBc), viral polymerase (Pol), and viral genome DNA. The 22 nm 5’ upstream region of the C gene and subsequent cleavage of the protein
particles, which are much more abundant in patient serum, include product at its C-terminus. Pol is the largest HBV protein, consisting of
subviral particles (SVPs) that lack the nucleocapsid and are thus four domains with three discrete enzymatic functions: 1) the terminal
non-infectious. Also, other non-infectious particles are currently known protein (TP) domain, which is essential for binding to pgRNA and serves
to be produced by infection, including enveloped particles that lack a as a protein primer to initiate minus strand DNA synthesis; 2) the spacer
viral genome, those containing viral RNA, and envelope-less particles

* Corresponding author. Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
E-mail address: kwatashi@nih.go.jp (K. Watashi).

https://doi.org/10.1016/j.antiviral.2020.104925
Received 4 May 2020; Received in revised form 24 July 2020; Accepted 26 August 2020
Available online 28 August 2020
0166-3542/© 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Abbreviations NTCP sodium taurocholate cotransporting peptide


p22cr 22-kDa precore protein
APOBEC3 apolipoprotein B editing complex 3 PAPD poly(A) RNA polymerase associated domain containing
CBP CREB-binding protein proteins
cccDNA covalently closed circular DNA PCAF CBP-associated factor
C/EBP CCAAT-enhancer-binding protein pgRNA pregenomic RNA
CREB cAMP response element binding protein Pol polymerase
DHBV duck hepatitis B virus Pol α DNA polymerase α
DR1 direct repeat 1 Pol κ DNA polymerase κ
eIF eukaryotic translation initiation factor PPARα peroxisome proliferator-activated receptor alpha
ESCRT endosomal sorting complex required for transport Prdx1 peroxiredoxin 1
FEN1 flap structure-specific endonuclease 1 PRMT protein methyl transferase
FXR farnesoid X receptor PROX1 prosperous-related homeobox protein 1
HBc hepatitis B virus core antigen RBM24 RNA-binding motif protein 24
HBe hepatitis B virus E antigen RBP24 RNA-binding motif protein 24
HBs hepatitis B virus surface antigen rcDNA relaxed-circular DNA
HBV hepatitis B virus RNaseH ribonuclease H
HDAC histone deacetylase RT reverse transcriptase
HNF hepatocyte nuclear factor RXR retinoid X receptor
HP1 heterochromatin protein 1 Set1A histone methyltransferase 1A
Hsc heat stress cognate SETDB1 SET domain bifurcated histone lysine methyltransferase 1
Hsp heat shock protein S-HBs small surface antigen
HSPGs heparan sulfate proteoglycans SIRT sirtuin
IFN interferon SKIV2L superkiller viralicidic activity 2-like
ISG20 IFN-stimulated exoribonuclease gene of 20 kDa Smc structural maintenance of chromosomes
L-HBs large surface antigen SP1 specificity protein 1
LIG DNA ligase SVPs subviral particles
LSD1 histone lysine demethylase 1 TDP2 tyrosyl-DNA phosphodiesterase 2
LRH-1 liver receptor homolog-1 TNF tumor necrosis factor
LTβ lymphotoxin β TOP topoisomerase
m6A N6-methyladenosine TP terminal protein
M-HBs middle surface antigen UBE2L3 ubiquitin conjugating enzyme E2 L3
MSL2 male-specific lethal 2 ZAP zinc finger antiviral protein
MVB multivesicular body ZHX2 zinc finger and homeoboxes 2
NF1 nuclear factor 1

domain, whose function has not been well defined; 3) the reverse expressed in the liver and originally functions to uptake bile salts into
transcriptase (RT) domain, which has DNA elongation activity for both hepatocytes, was identified as an HBV and HDV entry receptor in 2012.
reverse transcription and DNA-dependent DNA polymerization; and 4) NTCP was identified as a factor that bound to aa 2–48 of the preS1 re­
the ribonuclease H (RNaseH) domain, which digests pgRNA after reverse gion, which had been already known to be essential for receptor binding
transcription. The three HBs proteins share the common C-terminal S (Barrera et al., 2005; Glebe et al., 2005; Gripon et al., 2005; Ni et al.,
region. In addition, M-HBs and L-HBs carry an extended region, called 2014; Yan et al., 2012). Virus-receptor interactions are believed to
the preS2 region, at the N-terminus of the S region. L-HBs further in­ trigger virus internalization into cells in an endocytosis-dependent
cludes an extension (preS1 region), which is essential for receptor manner (Hao et al., 2011; Huang et al., 2012; Macovei et al., 2010,
binding during virus entry, at the N-terminus of the preS2 and S regions. 2013; Yamada et al., 2012). A recent report found that a receptor
HBx is a multifunctional protein that promotes viral production at tyrosine kinase, epidermal growth factor receptor (EGFR), triggers the
multiple steps, including viral transcription and replication, and also internalization of HBV/HDV through its direct interaction with NTCP
plays a role in the development of HBV-related hepatocellular (Iwamoto et al., 2019, 2020). It has also been reported recently that
carcinoma. NTCP can be oligomerized, and the oligomerization status modulates the
HBV proliferates in host cells by taking advantage of these viral ability of NTCP to mediate viral internalization (Fukano et al., 2018).
proteins in concert with host-derived factors. As an introduction to HBV Internalization in vesicles is believed to induce fusion between the viral
biology, in this article we summarize and update what is currently envelope and the cell-derived vesicular membrane, although its mech­
known about the mechanism of the HBV life cycle (Fig. 3). anism remains largely unknown. The incoming nucleocapsid in the
cytoplasm is directed to the nucleus along with the microtubules (Rabe
2. HBV entry et al., 2006) and is imported into the nucleus through the nuclear pore
complex in an importin-dependent manner (Kann et al., 1999; Rabe
HBV entry into host hepatocytes follows multiple steps. Initially, the et al., 2003).
virus attaches to the host cell surface through binding to factors
including heparan sulfate proteoglycans (HSPGs) such as glypican 5 in a 3. cccDNA formation/maintenance
non-specific and low-affinity manner, and then interacts with its re­
ceptor(s) more specifically and with high affinity (Leistner et al., 2008; In the nucleus, HBV genomic DNA is modified by cellular factors. The
Schulze et al., 2007; Verrier et al., 2016). Sodium taurocholate Pol-linked terminal redundant sequence in the 5′ -end of the minus
cotransporting peptide (NTCP/SLC10A1), which is specifically strand DNA and the RNA oligonucleotide attached at the 5′ end of the

2
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

plus strand DNA (see below for rcDNA structure and synthesis) are (MSL2) modulate cccDNA stability through degradation of APOBEC3A
removed from the rcDNA, and the gaps in both strands are filled and and APOBEC3B, respectively (Gao et al., 2017; Zhou et al., 2019). The
ligated to generate cccDNA (Gerlich and Robinson, 1980; Guo et al., ability of APOBEC3G to induce cccDNA hypermutation has also been
2007; Molnar-Kimber et al., 1983; Wang and Seeger, 1992). Several reported in an overexpression experiment (Kitamura et al., 2013). In
factors have been reported to be involved in the process of cccDNA addition to cccDNA formation and maintenance, a part of the incoming
formation. A DNA repair enzyme, tyrosyl-DNA phosphodiesterase 2 HBV DNA is integrated into the host genome. A recent cell culture study
(TDP2), has been shown to cleave the tyrosyl-DNA phosphodiester bond showed that the integration occurs as fast as within a week after infec­
between Pol and rcDNA in an in vitro assay, although its relevance in tion (Tu et al., 2018). The integrated HBV DNA is
cccDNA formation in the cellular context remains controversial (Cui replication-incompetent but can act as a template for the production of
et al., 2015; Koniger et al., 2014). Flap structure-specific endonuclease 1 HBs, which is thought to be related to HBV-specific immune tolerance
(FEN1), which is implicated in cellular DNA replication and repair, is and the development of HBV-related pathogenesis.
another factor reported to cleave the flap structure at the 5′ -end of the
minus strand (Kitamura et al., 2018). After the removal of the Pol and 4. HBV transcription
RNA primers, DNA polymerases and ligases, such as DNA polymerase κ
and α (Pol κ and Pol α), DNA ligase 1 and 3 (LIG1 and LIG3), and Using cccDNA as a template, four different lengths of RNAs (3.5, 2.4,
topoisomerase I and II (TOP1 and TOP2), have been reported to function 2.1, and 0.7 kb) are transcribed. Transcription of viral RNAs is regulated
for filling the gaps in rcDNA (Long et al., 2017; Qi et al., 2016; Sheraz by four distinct promoters for preS1, preS2, core, and X, and two en­
et al., 2019; Tang et al., 2019). Although it remains unknown how and hancers (Enhancer I and Enhancer II), which is mediated by host RNA
where cccDNA is maintained in the nucleus, cccDNA resides episomally polymerase II machinery-dependent transcription (Rall et al., 1983).
but is inherently stable, which thus functions as a template for viral The transcription is regulated at multiple levels as shown below (Fig. 4).
replication over the long term. A recent analysis showed a cccDNA
half-life of about 40 days in NTCP-overexpressing HepG2 cells (Ko et al., 4.1. Epigenetic modification
2018). But that in a clinical setting is likely to be far longer. A report
estimated more than 9 months for a cccDNA half-life in hepatitis B pa­ cccDNA exists as a minichromosome that associates with viral pro­
tients (Boyd et al., 2016). Immune responses and cytokine stimulation teins and host factors (Fig. 4). As cccDNA is assembled with histones, the
are major factors that affect cccDNA maintenance (Hu et al., 2019). The post-translational modification status of histones governs the tran­
mechanisms for the regulation of cccDNA maintenance/stability remain scriptional activity of cccDNA (Pollicino et al., 2006). Genome-wide
largely unclarified, but a cytidine deaminase, apolipoprotein B editing ChIP-seq analysis has revealed the post-translational modification of
complex 3 (APOBEC3), is an example of a protein that modulates cccDNA-associated histones that show a high level of trimethylation or
cccDNA stability. Upon stimulation by cytokines such as interferon α acetylation of lysine of histone 3 (H3K4me3, H3K27ac, and H3K122ac),
and γ (IFNα, IFNγ), tumor necrosis factor α (TNFα) as well as lympho­ active markers of transcription, enriched at specific sites within the HBV
toxin β (LTβ), APOBEC3A and/or APOBEC3B are induced and are re­ genome, and very low levels of transcriptional repression markers
ported to destabilize cccDNA (Lucifora et al., 2014; Xia et al., 2016). A (H3K27ac and H3K9me2) even at silent HBV promoters (Tropberger
ubiquitin conjugating enzyme E2 L3 (UBE2L3) and male-specific lethal 2 et al., 2015). There are also accumulating reports of histone

Fig. 1. Schematic representation of HBV particles. Infectious HBV virion (Dane particle) (upper) and non-infectious HBV particles, including enveloped capsids
containing immature DNA/RNA, subviral particles (sphere and filament), and naked nucleocapsids (lower).

3
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

modification enzymes and other factors directly or indirectly recruited 4.3. Role of HBx in HBV transcription
onto the cccDNA minichromosome to regulate viral transcription. Such
histone modification enzymes includes histone acetyltransferases HBx has been demonstrated to be essential for HBV replication after
[CREB-binding protein (CBP)/p300, CBP-associated factor infection (Lucifora et al., 2011). HBx is a multifunctional protein with
(PCAF)/CGN5, HAT1], histone deacetylases [histone deacetylase 1 and many studies showing a wide variety of functions, although the rele­
4 (HDAC1, HDAC4), sirtuin 1 and 3 (SIRT1, SIRT3)], methyltransferases vance of such functions in physiological settings needs to be further
[SET domain bifurcated histone lysine methyltransferase 1 (SETDB1), clarified. HBx has been reported to associate with the cccDNA mini­
protein methyl transferase 1 and 5 (PRMT1, PRMT5), histone methyl­ chromosome in close parallel to the kinetics of cccDNA-bound H3
transferase 1A (Set1A)], and demethylases [histone lysine demethylase acetylation (Belloni et al., 2009). HBx modulates the recruitment of
1 (LSD1)] (Alarcon et al., 2016; Belloni et al., 2009; Ren et al., 2018; chromatin-modifying enzymes (p300, HDAC, SIRT1) and controls the
Riviere et al., 2015; Xing et al., 2019; Yang et al., 2019; Zhang et al., epigenetic status of cccDNA-associated histones for active transcription
2017). More precise mechanisms for cccDNA-associated histone modi­ (Belloni et al., 2009). HBx has also been reported to affect not only
fication and its regulation of viral transcription are mentioned elsewhere acetylation but also methylation and phosphorylation of
(Hong et al., 2017). Importantly, these cccDNA-associated histone cccDNA-associated histones in HepG2 cells (Luo et al., 2013). In the
modifications have been revealed to be targeted by the antiviral activity absence of HBx, cccDNA is transcriptionally silenced by a decrease in H3
of interferon (IFN): IFNα induces hypoacetylation of cccDNA-bound acetylation and H3K4me3 and an increase in H3K9me2/3 that results in
histones and the recruitment of transcriptional corepressors that result the recruitment of heterochromatin protein 1 (HP1) and chromatin
in transcriptional silencing both in cell culture and in chimeric mouse condensation, while HBx expression relieves this transcriptional
models (Belloni et al., 2012). It has been also reported that IFNα alters silencing by recovering the increased H3K4me3 and dissociating HP1
the acetylation status of H3K9 and H3K27 to suppress transcription of recruitment on cccDNA (Riviere et al., 2015). This evidence supports the
duck hepatitis B virus (DHBV) cccDNA (Liu et al., 2013). activity of HBx on the epigenetic profile of cccDNA-associated histones
to regulate HBV transcription.
4.2. Transcription factors In addition, a recent finding on HBx action on Smc5/6 has opened a
new aspect of its function (Fig. 4). The structural maintenance of chro­
In addition to epigenetic control, the recruitment of cellular tran­ mosomes (Smc) family are ATPases that generally regulate higher-order
scription factors to the viral promoter/enhancer regions in cccDNA chromosome organization (Palecek, 2018). Recently, it has been
governs the activity of transcription. The viral promoter/enhancer re­ revealed that the Smc5/6 complex associates with an episomal HBV
gions contain the binding sites for numerous transcription factors, DNA reporter and suppresses its transcriptional activity (Decorsiere
including the elements for the liver-specific nuclear receptors. The et al., 2016). In the presence of HBx, DDB1-containing E3 ubiquitin
transcription factors involved in HBV transcription have been studied ligase is recruited to Smc5/6 and induces complex degradation to relieve
for more than two decades (Fig. 4). These include liver-enriched hepa­ Smc5/6-mediated transcriptional silencing. Genetic knockdown of
tocyte nuclear factor 3 and 4 (HNF3, HNF4), retinoid X receptor alpha Smc5/6 causes the replication of HBx-deficient HBV, suggesting an
(RXRα), peroxisome proliferator-activated receptor alpha (PPARα), and essential role for Smc5/6 antagonism in HBx’s function to support viral
farnesoid X receptor (FXR) as nuclear receptors (Chen et al., 1994; replication (Murphy et al., 2016). This function requires a CCCH motif in
Guidotti et al., 1999; Huan et al., 1995; Li et al., 1995; Ori and Shaul, the HBx sequence that is highly conserved among strains (Ramakrishnan
1995; Reese et al., 2013; Tang and McLachlan, 2001, 2002; Wang et al., et al., 2019). A study using clinical samples has shown that the
1998). Other transcription factors that have been reported to activate anti-Smc5/6 function can be retained in HBx variants found in HCC
transcription to augment pgRNA expression include patients (Riviere et al., 2019). Interestingly, nitazoxanide has been
CCAAT-enhancer-binding protein (C/EBP), nuclear factor 1 (NF1), found to inhibit HBx-DDB1 binding and restore the expression of Smc5
specificity protein 1 (SP1), cAMP response element binding protein to suppress HBV replication in HBV-infected cells, proposing that this
(CREB), and liver receptor homolog-1 (LRH-1), and those that report­ mechanism can serve as a therapeutic target (Sekiba et al., 2019).
edly suppress pgRNA transcription include HNF6, prosperous-related
homeobox protein 1 (PROX1), p53, and zinc finger and homeoboxes 2 5. HBV RNAs stability
(ZHX2) (Cai et al., 2003; Hao et al., 2015; Kim et al., 2008; Lopez-­
Cabrera et al., 1991; Ori et al., 1994; Qin et al., 2009; Uchida et al., Recent accumulating evidence has focused on the HBV RNA stability
1996; Xu et al., 2018; Yuh and Ting, 1991; Zhang and McLachlan, 1994; as a major step that limits the viral replication level. HBV pgRNA con­
Zhang et al., 1993). tains a stem loop, called epsilon, at both the 3’and 5′ termini. Epsilon is

Fig. 2. Schematic representation of the structure of HBV genomic DNA, RNAs, and proteins. Left, HBV genomic rcDNA and the encoded ORFs (C, P, S, X). Center,
HBV RNAs (gray lines) produced by cccDNA transcription and the proteins (boxes) produced from the RNAs. The RNA lengths, as well as the names of the RNAs, are
shown on the left. Right, HBV proteins and the domain structures. Amino acid numbers and the lengths are shown above the box and on the right, respectively.

4
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Fig. 3. Schematic overview of the HBV life cycle. See the main text for a precise description of the HBV life cycle.

Fig. 4. Schematic representation of the regulation of HBV transcription. HBV transcription is regulated at the epigenetic (left), transcription factor (center), and the
restriction (right) levels. Left, histone modification enzymes that positively (upper) and negatively (lower) regulate transcription are shown. Center, transcription
factors positively (upper) and negatively (lower) regulate the transcription of HBV core promoter/enhancer. Right, SMC5/6 suppresses the transcription, and HBx
counteracts the SMC5/6 restriction by the recruitment of the DDB1/Cul4 ubiquitin ligase complex for protein degradation.

5
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

essential for the RNA packaging into capsids (see below), and recent RNA at its 3′ epsilon stem loop structure, destabilizes HBV RNAs,
reports have shown a role for these elements in modulating the stability although the m6A at the 5′ epsilon stem loop of pgRNA has another
of HBV RNAs. Zinc finger antiviral protein (ZAP) interacts with HBV function that facilitates the reverse transcription of pgRNA (Imam et al.,
RNAs through a region containing the epsilon and promotes its decay 2018). m6A-modified HBV RNA at A1907 is recognized by YTH-domain
mainly in the nucleus, and this mechanism is potentiated upon IFN family 2 (YTHDF2) and an IFN-induced RNase, ISG20, which then
treatment (Mao et al., 2013). An RNA helicase, superkiller viralicidic processes HBV RNA for degradation (Imam et al., 2020). Thus, HBV RNA
activity 2-like (SKIV2L: a homolog of the Saccharomyces cerevisiae Ski2), stability has been revealed to be strictly regulated by multiple cellular
interacts with HBV RNAs, especially X-mRNA (0.7 Kb RNA), and me­ factors.
diates their degradation through a non-stop-mediated RNA decay
mechanism (Aly et al., 2016). A ribonuclease, IFN-stimulated exoribo­ 6. Encapsidation and DNA synthesis
nuclease gene of 20 kDa (ISG20), promotes HBV RNA degradation by
binding to the lower stem portion of the epsilon (Leong et al., 2016; Liu RNA encapsidation requires HBc, Pol, and viral RNA. Synthesized
et al., 2017). The growing list of other cellular factors that bind to HBV HBc monomers initially associate to yield a dimer, and then 90 or 120
RNAs and promote their degradation includes a cytidine deaminase, AID dimers subsequently self-assemble to constitute an icosahedral capsid
(Liang et al., 2015), a splicing factor and a U2 small nuclear ribonu­ (Bottcher et al., 1997; Conway et al., 1997). Concomitantly with the
cleoprotein auxiliary factor, PUF60 (Sun et al., 2017), RNA-binding encapsidation, Pol at its TP domain interacts with the epsilon stem loop
motif protein 24 (RBP24) (Yao et al., 2018), and peroxiredoxin 1 of the pgRNA at the 5’ terminus to form a ribonucleoprotein complex,
(Prdx1) (Deng et al., 2019). Interestingly, using a dihydroquinolizinone which is then incorporated into the capsid (Bartenschlager and Schaller,
derivative, RG7834, which is a new drug candidate that suppresses HBV 1992; Pollack and Ganem, 1994). This process is facilitated by host
replication and gene expression, the non-canonical poly(A) RNA poly­ chaperones, heat shock protein 90 (Hsp90), Hsp40, and heat stress
merase associated domain containing proteins 5 and 7 (PAPD5 and cognate 70 (Hsc70), through interactions with Pol and its conforma­
PAPD7) has been revealed to function in stabilizing HBV RNA, and tional optimization(Beck and Nassal, 2003; Hu et al., 1997, 2004; Hu
RG7834 has been proved to target these proteins (Mueller et al., 2018, and Seeger, 1996; Prange et al., 1999; Stahl et al., 2007). RNA-binding
2019). It has also been recently reported that covalent modifications of motif protein 24 (RBM24) interacts with both Pol and epsilon RNA to
viral RNA can regulate the translation and/or stability of RNA in many mediate the encapsidation (Yao et al., 2019). The interaction of nucle­
viruses including HIV-1, influenza virus, enterovirus, and respiratory ophosmin B23 with HBc also increases capsid assembly (Jeong et al.,
syncytial virus, which is known as epitranscriptomic regulation (Tsai 2014; Lee et al., 2009). The reported host factors incorporated into the
and Cullen, 2020). In HBV, it has been reported that the capsid include eukaryotic translation initiation factor 4E (eIF4E),
post-transcriptional modification, N6-methyladenosine (m6A) of HBV DEAD-box RNA helicase DDX3, and APOBEC3G (Kim et al., 2010;

Fig. 5. Schematic representation of reverse transcription, RNA degradation, and DNA synthesis in nucleocapsids. Pol (shown by TP, RT, RH) is recruited to the 5′ -
terminal epsilon and mediates the priming reaction. After translocation to the 3′ -terminal DR1, Pol mediates the DNA elongation to synthesize the full-length (− )DNA
and simultaneously digests the template RNA. The remaining 5′ -terminal RNA fragment containing DR1 is translocated to DR2 on the other terminus and starts the
synthesis of (+)DNA.

6
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Nguyen and Hu, 2008; Turelli et al., 2004; Wang et al., 2009). DDX3 and Smoking Research Foundation; and the Mochida Memorial Foundation
APOBEC3G have a negative effect on the HBV replication. for Medical and Pharmaceutical Research. The authors declare that they
After RNA incorporation, viral genome synthesis proceeds through have no conflicts of interest with the contents of this article.
several steps inside the nucleocapsid (Fig. 5). The epsilon in pgRNA has
an internal bulge possessing the sequence 5′ -UUC-3′ , which functions as References
a template for priming. A tyrosine residue at aa 63 in the TP domain of
Pol acts as a protein primer: the first dGTP residue is covalently linked to Alarcon, V., Hernandez, S., Rubio, L., Alvarez, F., Flores, Y., Varas-Godoy, M., De
Ferrari, G.V., Kann, M., Villanueva, R.A., Loyola, A., 2016. The enzymes LSD1 and
a hydroxyl residue in Y63 (priming initiation), and two further dAMPs Set1A cooperate with the viral protein HBx to establish an active hepatitis B viral
are extended to produce 5′ -dGAA-3’ (priming polymerization) (Jones chromatin state. Sci. Rep. 6, 25901.
et al., 2012; Nassal and Rieger, 1996; Wang and Seeger, 1992, 1993; Aly, H.H., Suzuki, J., Watashi, K., Chayama, K., Hoshino, S., Hijikata, M., Kato, T.,
Wakita, T., 2016. RNA exosome complex regulates stability of the hepatitis B virus X-
Wang and Hu, 2002). The produced Pol-dGAA complex is then trans­ mRNA transcript in a non-stop-mediated (NSD) RNA quality control mechanism.
located to the complementary direct repeat 1 (DR1) sequence at the 3′ J. Biol. Chem. 291, 15958–15974.
end of pgRNA to allow for minus strand DNA synthesis (Lin et al., 2008; Bardens, A., Doring, T., Stieler, J., Prange, R., 2011. Alix regulates egress of hepatitis B
virus naked capsid particles in an ESCRT-independent manner. Cell Microbiol. 13,
Wang and Hu, 2002). This strand is extended to the 3′ end of pgRNA, 602–619.
resulting in a unit length minus strand DNA containing an additional Barrera, A., Guerra, B., Notvall, L., Lanford, R.E., 2005. Mapping of the hepatitis B virus
terminal redundancy. The pgRNA template is simultaneously degraded pre-S1 domain involved in receptor recognition. J. Virol. 79, 9786–9798.
Bartenschlager, R., Schaller, H., 1992. Hepadnaviral assembly is initiated by polymerase
during the minus strand DNA synthesis through digestion by the RNase
binding to the encapsidation signal in the viral RNA genome. EMBO J. 11,
H domain, and eventually leaves a short RNA fragment containing the 3413–3420.
capped 5′ terminal region including 5′ DR1. This RNA fragment is then Beck, J., Nassal, M., 2003. Efficient Hsp90-independent in vitro activation by Hsc70 and
translocated to DR2 at the 3′ terminus and is extended to the 5′ terminus Hsp40 of duck hepatitis B virus reverse transcriptase, an assumed Hsp90 client
protein. J. Biol. Chem. 278, 36128–36138.
of the minus strand DNA. Having a redundant sequence, extending the 3′ Belloni, L., Allweiss, L., Guerrieri, F., Pediconi, N., Volz, T., Pollicino, T., Petersen, J.,
end of the plus strand DNA switches to the 3′ redundant sequence on the Raimondo, G., Dandri, M., Levrero, M., 2012. IFN-alpha inhibits HBV transcription
minus strand DNA, enabling further elongation of the plus strand DNA to and replication in cell culture and in humanized mice by targeting the epigenetic
regulation of the nuclear cccDNA minichromosome. J. Clin. Invest. 122, 529–537.
eventually produce rcDNA. Belloni, L., Pollicino, T., De Nicola, F., Guerrieri, F., Raffa, G., Fanciulli, M.,
Raimondo, G., Levrero, M., 2009. Nuclear HBx binds the HBV minichromosome and
7. HBV morphogenesis modifies the epigenetic regulation of cccDNA function. Proc. Natl. Acad. Sci. U. S. A.
106, 19975–19979.
Blumberg, B.S., Alter, H.J., Visnich, S., 1965. A "new" antigen in leukemia sera. J. Am.
The viral genome-containing nucleocapsids are either shuttled to the Med. Assoc. 191, 541–546.
nucleus to amplify and maintain the cccDNA pool (recycling) or Bottcher, B., Wynne, S.A., Crowther, R.A., 1997. Determination of the fold of the core
protein of hepatitis B virus by electron cryomicroscopy. Nature 386, 88–91.
assembled with mature envelope proteins to be secreted outside of cells Boyd, A., Lacombe, K., Lavocat, F., Maylin, S., Miailhes, P., Lascoux-Combe, C.,
(virus egress). Capsids containing the replication intermediates, pgRNA, Delaugerre, C., Girard, P.M., Zoulim, F., 2016. Decay of ccc-DNA marks persistence
and those without viral DNA/RNA are also secreted from cells but with of intrahepatic viral DNA synthesis under tenofovir in HIV-HBV co-infected patients.
J. Hepatol. 65, 683–691.
different secretion efficiencies. The secretion process for infectious vi­
Cai, Y.N., Zhou, Q., Kong, Y.Y., Li, M., Viollet, B., Xie, Y.H., Wang, Y., 2003. LRH-1/hB1F
rions depends on multivesicular body (MVB)-associated endosomal and HNF1 synergistically up-regulate hepatitis B virus gene transcription and DNA
sorting complex required for transport (ESCRT) machinery, which in­ replication. Cell Res. 13, 451–458.
Chen, M., Hieng, S., Qian, X., Costa, R., Ou, J.H., 1994. Regulation of hepatitis B virus
volves gamma 2-adaptin, CHMP3/4, Vps4, Nedd4, and α-taxilin (Hoff­
ENI enhancer activity by hepatocyte-enriched transcription factor HNF3. Virology
mann et al., 2013; Lambert et al., 2007; Rost et al., 2006, 2008; Stieler 205, 127–132.
and Prange, 2014; Watanabe et al., 2007). In contrast, the egress of Chou, S.F., Tsai, M.L., Huang, J.Y., Chang, Y.S., Shih, C., 2015. The dual role of an
naked capsids does not require the above ESCRT machinery, but rather ESCRT-0 component HGS in HBV transcription and naked capsid secretion. PLoS
Pathog. 11, e1005123.
involves other factors such as Alix and HGS (Bardens et al., 2011; Chou Conway, J.F., Cheng, N., Zlotnick, A., Wingfield, P.T., Stahl, S.J., Steven, A.C., 1997.
et al., 2015). Subviral particles in the spherical structure that predom­ Visualization of a 4-helix bundle in the hepatitis B virus capsid by cryo-electron
inantly contain S-HBs are released from the endoplasmic reticulum microscopy. Nature 386, 91–94.
Cui, X., McAllister, R., Boregowda, R., Sohn, J.A., Cortes Ledesma, F., Caldecott, K.W.,
through the general secretory pathway (Patient et al., 2007, 2009), and Seeger, C., Hu, J., 2015. Does tyrosyl DNA phosphodiesterase-2 play a role in
filamentous subviral particles that also contain L-HBs share the hepatitis B virus genome repair? PloS One 10, e0128401.
ESCRT-dependent egress pathway (Jiang et al., 2015). Thus, the virus Dane, D.S., Cameron, C.H., Briggs, M., 1970. Virus-like particles in serum of patients with
Australia-antigen-associated hepatitis. Lancet 1, 695–698.
egress pathway is closely associated with the cellular membrane sorting Decorsiere, A., Mueller, H., van Breugel, P.C., Abdul, F., Gerossier, L., Beran, R.K.,
machinery. Livingston, C.M., Niu, C., Fletcher, S.P., Hantz, O., Strubin, M., 2016. Hepatitis B
virus X protein identifies the Smc5/6 complex as a host restriction factor. Nature
531, 386–389.
8. Conclusions
Deng, L., Gan, X., Ito, M., Chen, M., Aly, H.H., Matsui, C., Abe, T., Watashi, K.,
Wakita, T., Suzuki, T., Okamoto, T., Matsuura, Y., Mizokami, M., Shoji, I., Hotta, H.,
Numerous studies have been conducted to better comprehend HBV 2019. Peroxiredoxin 1, a novel HBx-interacting protein, interacts with exosome
component 5 and negatively regulates hepatitis B virus (HBV) propagation through
biology. This has led to the identification of numerous targets that are
degradation of HBV RNA. J. Virol. 93.
currently under clinical and preclinical development (see other articles Fukano, K., Tsukuda, S., Oshima, M., Suzuki, R., Aizaki, H., Ohki, M., Park, S.Y.,
in this issue). Further clarification will provide additional anti-HBV drug Muramatsu, M., Wakita, T., Sureau, C., Ogasawara, Y., Watashi, K., 2018.
candidates with novel modes of action. Troglitazone impedes the oligomerization of sodium taurocholate cotransporting
polypeptide and entry of hepatitis B virus into hepatocytes. Front. Microbiol. 9,
3257.
Acknowledgments Gao, Y., Feng, J., Yang, G., Zhang, S., Liu, Y., Bu, Y., Sun, M., Zhao, M., Chen, F.,
Zhang, W., Ye, L., Zhang, X., 2017. Hepatitis B virus X protein-elevated MSL2
modulates hepatitis B virus covalently closed circular DNA by inducing degradation
We appreciate Drs. Julie Lucifora and Marion Delphin at INSERM, of APOBEC3B to enhance hepatocarcinogenesis. Hepatology 66, 1413–1429.
U1052, Université de Lyon for their scientific comments. The authors’ Gerlich, W.H., Robinson, W.S., 1980. Hepatitis B virus contains protein attached to the 5’
work was supported by the Japan Society for the Promotion of Science terminus of its complete DNA strand. Cell 21, 801–809.
Glebe, D., Urban, S., Knoop, E.V., Cag, N., Krass, P., Grun, S., Bulavaite, A.,
KAKENHI Grants JP17H04085 and JP20H03499; the Japan Science and Sasnauskas, K., Gerlich, W.H., 2005. Mapping of the hepatitis B virus attachment site
Technology Agency (JST) MIRAI program; the Japan Agency for Medi­ by use of infection-inhibiting preS1 lipopeptides and tupaia hepatocytes.
cal Research and Development (AMED) Grants JP20fk0310114j0004, Gastroenterology 129, 234–245.
JP20fk0310101j1004, JP20fk0310103j0204, JP20fk0210036j0003,
and JP20jm0210068j0002; the Takeda Science Foundation; the

7
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Gripon, P., Cannie, I., Urban, S., 2005. Efficient inhibition of hepatitis B virus infection Koniger, C., Wingert, I., Marsmann, M., Rosler, C., Beck, J., Nassal, M., 2014.
by acylated peptides derived from the large viral surface protein. J. Virol. 79, Involvement of the host DNA-repair enzyme TDP2 in formation of the covalently
1613–1622. closed circular DNA persistence reservoir of hepatitis B viruses. Proc. Natl. Acad. Sci.
Guidotti, L.G., Eggers, C.M., Raney, A.K., Chi, S.Y., Peters, J.M., Gonzalez, F.J., U. S. A. 111, E4244–E4253.
McLachlan, A., 1999. In vivo regulation of hepatitis B virus replication by Lambert, C., Doring, T., Prange, R., 2007. Hepatitis B virus maturation is sensitive to
peroxisome proliferators. J. Virol. 73, 10377–10386. functional inhibition of ESCRT-III, Vps4, and gamma 2-adaptin. J. Virol. 81,
Guo, H., Jiang, D., Zhou, T., Cuconati, A., Block, T.M., Guo, J.T., 2007. Characterization 9050–9060.
of the intracellular deproteinized relaxed circular DNA of hepatitis B virus: an Lee, S.J., Shim, H.Y., Hsieh, A., Min, J.Y., Jung, G., 2009. Hepatitis B virus core interacts
intermediate of covalently closed circular DNA formation. J. Virol. 81, with the host cell nucleolar protein, nucleophosmin 1. J. Microbiol. 47, 746–752.
12472–12484. Leistner, C.M., Gruen-Bernhard, S., Glebe, D., 2008. Role of glycosaminoglycans for
Hao, R., He, J., Liu, X., Gao, G., Liu, D., Cui, L., Yu, G., Yu, W., Chen, Y., Guo, D., 2015. binding and infection of hepatitis B virus. Cell Microbiol. 10, 122–133.
Inhibition of hepatitis B virus gene expression and replication by hepatocyte nuclear Leong, C.R., Funami, K., Oshiumi, H., Mengao, D., Takaki, H., Matsumoto, M., Aly, H.H.,
factor 6. J. Virol. 89, 4345–4355. Watashi, K., Chayama, K., Seya, T., 2016. Interferon-stimulated gene of 20 kDa
Hao, X., Shang, X., Wu, J., Shan, Y., Cai, M., Jiang, J., Huang, Z., Tang, Z., Wang, H., protein (ISG20) degrades RNA of hepatitis B virus to impede the replication of HBV
2011. Single-particle tracking of hepatitis B virus-like vesicle entry into cells. Small in vitro and in vivo. Oncotarget 7, 68179–68193.
7, 1212–1218. Li, M., Xie, Y., Wu, X., Kong, Y., Wang, Y., 1995. HNF3 binds and activates the second
Hoffmann, J., Boehm, C., Himmelsbach, K., Donnerhak, C., Roettger, H., Weiss, T.S., enhancer, ENII, of hepatitis B virus. Virology 214, 371–378.
Ploen, D., Hildt, E., 2013. Identification of alpha-taxilin as an essential factor for the Liang, G., Liu, G., Kitamura, K., Wang, Z., Chowdhury, S., Monjurul, A.M., Wakae, K.,
life cycle of hepatitis B virus. J. Hepatol. 59, 934–941. Koura, M., Shimadu, M., Kinoshita, K., Muramatsu, M., 2015. TGF-beta suppression
Hong, X., Kim, E.S., Guo, H., 2017. Epigenetic regulation of hepatitis B virus covalently of HBV RNA through AID-dependent recruitment of an RNA exosome complex. PLoS
closed circular DNA: implications for epigenetic therapy against chronic hepatitis B. Pathog. 11, e1004780.
Hepatology 66, 2066–2077. Lin, L., Wan, F., Hu, J., 2008. Functional and structural dynamics of hepadnavirus
Hu, J., Flores, D., Toft, D., Wang, X., Nguyen, D., 2004. Requirement of heat shock reverse transcriptase during protein-primed initiation of reverse transcription:
protein 90 for human hepatitis B virus reverse transcriptase function. J. Virol. 78, effects of metal ions. J. Virol. 82, 5703–5714.
13122–13131. Liu, F., Campagna, M., Qi, Y., Zhao, X., Guo, F., Xu, C., Li, S., Li, W., Block, T.M.,
Hu, J., Liu, K., 2017. Complete and incomplete hepatitis B virus particles: formation, Chang, J., Guo, J.T., 2013. Alpha-interferon suppresses hepadnavirus transcription
function, and application. Viruses 9. by altering epigenetic modification of cccDNA minichromosomes. PLoS Pathog. 9,
Hu, J., Protzer, U., Siddiqui, A., 2019. Revisiting hepatitis B virus: challenges of curative e1003613.
therapies. J. Virol. 93. Liu, Y., Nie, H., Mao, R., Mitra, B., Cai, D., Yan, R., Guo, J.T., Block, T.M., Mechti, N.,
Hu, J., Seeger, C., 1996. Hsp90 is required for the activity of a hepatitis B virus reverse Guo, H., 2017. Interferon-inducible ribonuclease ISG20 inhibits hepatitis B virus
transcriptase. Proc. Natl. Acad. Sci. U. S. A. 93, 1060–1064. replication through directly binding to the epsilon stem-loop structure of viral RNA.
Hu, J., Toft, D.O., Seeger, C., 1997. Hepadnavirus assembly and reverse transcription PLoS Pathog. 13, e1006296.
require a multi-component chaperone complex which is incorporated into Long, Q., Yan, R., Hu, J., Cai, D., Mitra, B., Kim, E.S., Marchetti, A., Zhang, H., Wang, S.,
nucleocapsids. EMBO J. 16, 59–68. Liu, Y., Huang, A., Guo, H., 2017. The role of host DNA ligases in hepadnavirus
Huan, B., Kosovsky, M.J., Siddiqui, A., 1995. Retinoid X receptor alpha transactivates the covalently closed circular DNA formation. PLoS Pathog. 13, e1006784.
hepatitis B virus enhancer 1 element by forming a heterodimeric complex with the Lopez-Cabrera, M., Letovsky, J., Hu, K.Q., Siddiqui, A., 1991. Transcriptional factor C/
peroxisome proliferator-activated receptor. J. Virol. 69, 547–551. EBP binds to and transactivates the enhancer element II of the hepatitis B virus.
Huang, H.C., Chen, C.C., Chang, W.C., Tao, M.H., Huang, C., 2012. Entry of hepatitis B Virology 183, 825–829.
virus into immortalized human primary hepatocytes by clathrin-dependent Lucifora, J., Arzberger, S., Durantel, D., Belloni, L., Strubin, M., Levrero, M., Zoulim, F.,
endocytosis. J. Virol. 86, 9443–9453. Hantz, O., Protzer, U., 2011. Hepatitis B virus X protein is essential to initiate and
Imam, H., Khan, M., Gokhale, N.S., McIntyre, A.B.R., Kim, G.W., Jang, J.Y., Kim, S.J., maintain virus replication after infection. J. Hepatol. 55, 996–1003.
Mason, C.E., Horner, S.M., Siddiqui, A., 2018. N6-methyladenosine modification of Lucifora, J., Xia, Y., Reisinger, F., Zhang, K., Stadler, D., Cheng, X., Sprinzl, M.F.,
hepatitis B virus RNA differentially regulates the viral life cycle. Proc. Natl. Acad. Koppensteiner, H., Makowska, Z., Volz, T., Remouchamps, C., Chou, W.M.,
Sci. U. S. A. 115, 8829–8834. Thasler, W.E., Huser, N., Durantel, D., Liang, T.J., Munk, C., Heim, M.H.,
Imam, H., Kim, G.W., Mir, S.A., Khan, M., Siddiqui, A., 2020. Interferon-stimulated gene Browning, J.L., Dejardin, E., Dandri, M., Schindler, M., Heikenwalder, M.,
20 (ISG20) selectively degrades N6-methyladenosine modified Hepatitis B Virus Protzer, U., 2014. Specific and nonhepatotoxic degradation of nuclear hepatitis B
transcripts. PLoS Pathog. 16, e1008338. virus cccDNA. Science 343, 1221–1228.
Iwamoto, M., Saso, W., Nishioka, K., Ohashi, H., Sugiyama, R., Ryo, A., Ohki, M., Yun, J. Luo, L., Chen, S., Gong, Q., Luo, N., Lei, Y., Guo, J., He, S., 2013. Hepatitis B virus X
H., Park, S.Y., Ohshima, T., Suzuki, R., Aizaki, H., Muramatsu, M., Matano, T., protein modulates remodelling of minichromosomes related to hepatitis B virus
Iwami, S., Sureau, C., Wakita, T., Watashi, K., 2020. The machinery for endocytosis replication in HepG2 cells. Int. J. Mol. Med. 31, 197–204.
of epidermal growth factor receptor coordinates the transport of incoming hepatitis Macovei, A., Petrareanu, C., Lazar, C., Florian, P., Branza-Nichita, N., 2013. Regulation
B virus to the endosomal network. J. Biol. Chem. 295, 800–807. of hepatitis B virus infection by Rab5, Rab7, and the endolysosomal compartment.
Iwamoto, M., Saso, W., Sugiyama, R., Ishii, K., Ohki, M., Nagamori, S., Suzuki, R., J. Virol. 87, 6415–6427.
Aizaki, H., Ryo, A., Yun, J.H., Park, S.Y., Ohtani, N., Muramatsu, M., Iwami, S., Macovei, A., Radulescu, C., Lazar, C., Petrescu, S., Durantel, D., Dwek, R.A.,
Tanaka, Y., Sureau, C., Wakita, T., Watashi, K., 2019. Epidermal growth factor Zitzmann, N., Nichita, N.B., 2010. Hepatitis B virus requires intact caveolin-1
receptor is a host-entry cofactor triggering hepatitis B virus internalization. Proc. function for productive infection in HepaRG cells. J. Virol. 84, 243–253.
Natl. Acad. Sci. U. S. A. 116, 8487–8492. Mao, R., Nie, H., Cai, D., Zhang, J., Liu, H., Yan, R., Cuconati, A., Block, T.M., Guo, J.T.,
Jeong, H., Cho, M.H., Park, S.G., Jung, G., 2014. Interaction between nucleophosmin and Guo, H., 2013. Inhibition of hepatitis B virus replication by the host zinc finger
HBV core protein increases HBV capsid assembly. FEBS Lett. 588, 851–858. antiviral protein. PLoS Pathog. 9, e1003494.
Jiang, B., Himmelsbach, K., Ren, H., Boller, K., Hildt, E., 2015. Subviral hepatitis B virus Molnar-Kimber, K.L., Summers, J., Taylor, J.M., Mason, W.S., 1983. Protein covalently
filaments, like infectious viral particles, are released via multivesicular bodies. bound to minus-strand DNA intermediates of duck hepatitis B virus. J. Virol. 45,
J. Virol. 90, 3330–3341. 165–172.
Jones, S.A., Boregowda, R., Spratt, T.E., Hu, J., 2012. In vitro epsilon RNA-dependent Mueller, H., Lopez, A., Tropberger, P., Wildum, S., Schmaler, J., Pedersen, L., Han, X.,
protein priming activity of human hepatitis B virus polymerase. J. Virol. 86, Wang, Y., Ottosen, S., Yang, S., Young, J.A.T., Javanbakht, H., 2019. PAPD5/7 are
5134–5150. host factors that are required for hepatitis B virus RNA stabilization. Hepatology 69,
Kann, M., Sodeik, B., Vlachou, A., Gerlich, W.H., Helenius, A., 1999. Phosphorylation- 1398–1411.
dependent binding of hepatitis B virus core particles to the nuclear pore complex. Mueller, H., Wildum, S., Luangsay, S., Walther, J., Lopez, A., Tropberger, P.,
J. Cell Biol. 145, 45–55. Ottaviani, G., Lu, W., Parrott, N.J., Zhang, J.D., Schmucki, R., Racek, T., Hoflack, J.
Kim, B.K., Lim, S.O., Park, Y.G., 2008. Requirement of the cyclic adenosine C., Kueng, E., Point, F., Zhou, X., Steiner, G., Lutgehetmann, M., Rapp, G., Volz, T.,
monophosphate response element-binding protein for hepatitis B virus replication. Dandri, M., Yang, S., Young, J.A.T., Javanbakht, H., 2018. A novel orally available
Hepatology 48, 361–373. small molecule that inhibits hepatitis B virus expression. J. Hepatol. 68, 412–420.
Kim, S., Wang, H., Ryu, W.S., 2010. Incorporation of eukaryotic translation initiation Murphy, C.M., Xu, Y., Li, F., Nio, K., Reszka-Blanco, N., Li, X., Wu, Y., Yu, Y., Xiong, Y.,
factor eIF4E into viral nucleocapsids via interaction with hepatitis B virus Su, L., 2016. Hepatitis B virus X protein promotes degradation of SMC5/6 to enhance
polymerase. J. Virol. 84, 52–58. HBV replication. Cell Rep. 16, 2846–2854.
Kitamura, K., Que, L., Shimadu, M., Koura, M., Ishihara, Y., Wakae, K., Nakamura, T., Nassal, M., Rieger, A., 1996. A bulged region of the hepatitis B virus RNA encapsidation
Watashi, K., Wakita, T., Muramatsu, M., 2018. Flap endonuclease 1 is involved in signal contains the replication origin for discontinuous first-strand DNA synthesis.
cccDNA formation in the hepatitis B virus. PLoS Pathog. 14, e1007124. J. Virol. 70, 2764–2773.
Kitamura, K., Wang, Z., Chowdhury, S., Simadu, M., Koura, M., Muramatsu, M., 2013. Nguyen, D.H., Hu, J., 2008. Reverse transcriptase- and RNA packaging signal-dependent
Uracil DNA glycosylase counteracts APOBEC3G-induced hypermutation of hepatitis incorporation of APOBEC3G into hepatitis B virus nucleocapsids. J. Virol. 82,
B viral genomes: excision repair of covalently closed circular DNA. PLoS Pathog. 9, 6852–6861.
e1003361. Ni, Y., Lempp, F.A., Mehrle, S., Nkongolo, S., Kaufman, C., Falth, M., Stindt, J.,
Ko, C., Chakraborty, A., Chou, W.M., Hasreiter, J., Wettengel, J.M., Stadler, D., Koniger, C., Nassal, M., Kubitz, R., Sultmann, H., Urban, S., 2014. Hepatitis B and D
Bester, R., Asen, T., Zhang, K., Wisskirchen, K., McKeating, J.A., Ryu, W.S., viruses exploit sodium taurocholate co-transporting polypeptide for species-specific
Protzer, U., 2018. Hepatitis B virus genome recycling and de novo secondary entry into hepatocytes. Gastroenterology 146, 1070–1083.
infection events maintain stable cccDNA levels. J. Hepatol. 69, 1231–1241. Okochi, K., Murakami, S., 1968. Observations on Australia antigen in Japanese. Vox
Sang. 15, 374–385.

8
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Ori, A., Atzmony, D., Haviv, I., Shaul, Y., 1994. An NF1 motif plays a central role in Tang, H., McLachlan, A., 2001. Transcriptional regulation of hepatitis B virus by nuclear
hepatitis B virus enhancer. Virology 204, 600–608. hormone receptors is a critical determinant of viral tropism. Proc. Natl. Acad. Sci. U.
Ori, A., Shaul, Y., 1995. Hepatitis B virus enhancer binds and is activated by the S. A. 98, 1841–1846.
Hepatocyte nuclear factor 3. Virology 207, 98–106. Tang, H., McLachlan, A., 2002. Mechanisms of inhibition of nuclear hormone receptor-
Palecek, J.J., 2018. SMC5/6: multifunctional player in replication. Genes 10. dependent hepatitis B virus replication by hepatocyte nuclear factor 3beta. J. Virol.
Patient, R., Hourioux, C., Roingeard, P., 2009. Morphogenesis of hepatitis B virus and its 76, 8572–8581.
subviral envelope particles. Cell Microbiol. 11, 1561–1570. Tang, L., Sheraz, M., McGrane, M., Chang, J., Guo, J.T., 2019. DNA Polymerase alpha is
Patient, R., Hourioux, C., Sizaret, P.Y., Trassard, S., Sureau, C., Roingeard, P., 2007. essential for intracellular amplification of hepatitis B virus covalently closed circular
Hepatitis B virus subviral envelope particle morphogenesis and intracellular DNA. PLoS Pathog. 15, e1007742.
trafficking. J. Virol. 81, 3842–3851. Tropberger, P., Mercier, A., Robinson, M., Zhong, W., Ganem, D.E., Holdorf, M., 2015.
Pollack, J.R., Ganem, D., 1994. Site-specific RNA binding by a hepatitis B virus reverse Mapping of histone modifications in episomal HBV cccDNA uncovers an unusual
transcriptase initiates two distinct reactions: RNA packaging and DNA synthesis. chromatin organization amenable to epigenetic manipulation. Proc. Natl. Acad. Sci.
J. Virol. 68, 5579–5587. U. S. A. 112, E5715–E5724.
Pollicino, T., Belloni, L., Raffa, G., Pediconi, N., Squadrito, G., Raimondo, G., Levrero, M., Tsai, K., Cullen, B.R., 2020. Epigenetic and epitranscriptomic regulation of viral
2006. Hepatitis B virus replication is regulated by the acetylation status of hepatitis replication. Nat. Rev. Microbiol. 1–12. https://doi.org/10.1038/s41579-020-0382-
B virus cccDNA-bound H3 and H4 histones. Gastroenterology 130, 823–837. 3.
Prange, R., Werr, M., Loffler-Mary, H., 1999. Chaperones involved in hepatitis B virus Tu, T., Budzinska, M.A., Vondran, F.W.R., Shackel, N.A., Urban, S., 2018. Hepatitis B
morphogenesis. Biol. Chem. 380, 305–314. virus DNA integration occurs early in the viral life cycle in an in vitro infection
Prince, A.M., 1968. An antigen detected in the blood during the incubation period of model via sodium taurocholate cotransporting polypeptide-dependent uptake of
serum hepatitis. Proc. Natl. Acad. Sci. U. S. A. 60, 814–821. enveloped virus particles. J. Virol. 92.
Qi, Y., Gao, Z., Xu, G., Peng, B., Liu, C., Yan, H., Yao, Q., Sun, G., Liu, Y., Tang, D., Turelli, P., Mangeat, B., Jost, S., Vianin, S., Trono, D., 2004. Inhibition of hepatitis B
Song, Z., He, W., Sun, Y., Guo, J.T., Li, W., 2016. DNA polymerase kappa is a key virus replication by APOBEC3G. Science 303, 1829.
cellular factor for the formation of covalently closed circular DNA of hepatitis B Uchida, T., Takahashi, K., Tatsuno, K., Dhingra, U., Eliason, J.F., 1996. Inhibition of
virus. PLoS Pathog. 12, e1005893. hepatitis-B-virus core promoter by p53: implications for carcinogenesis in
Qin, J., Zhai, J., Hong, R., Shan, S., Kong, Y., Wen, Y., Wang, Y., Liu, J., Xie, Y., 2009. hepatocytes. Int. J. Canc. 67, 892–897.
Prospero-related homeobox protein (Prox1) inhibits hepatitis B virus replication Verrier, E.R., Colpitts, C.C., Bach, C., Heydmann, L., Weiss, A., Renaud, M., Durand, S.C.,
through repressing multiple cis regulatory elements. J. Gen. Virol. 90, 1246–1255. Habersetzer, F., Durantel, D., Abou-Jaoude, G., Lopez Ledesma, M.M., Felmlee, D.J.,
Rabe, B., Glebe, D., Kann, M., 2006. Lipid-mediated introduction of hepatitis B virus Soumillon, M., Croonenborghs, T., Pochet, N., Nassal, M., Schuster, C., Brino, L.,
capsids into nonsusceptible cells allows highly efficient replication and facilitates the Sureau, C., Zeisel, M.B., Baumert, T.F., 2016. A targeted functional RNA interference
study of early infection events. J. Virol. 80, 5465–5473. screen uncovers glypican 5 as an entry factor for hepatitis B and D viruses.
Rabe, B., Vlachou, A., Pante, N., Helenius, A., Kann, M., 2003. Nuclear import of Hepatology 63, 35–48.
hepatitis B virus capsids and release of the viral genome. Proc. Natl. Acad. Sci. U. S. Wang, G.H., Seeger, C., 1992. The reverse transcriptase of hepatitis B virus acts as a
A. 100, 9849–9854. protein primer for viral DNA synthesis. Cell 71, 663–670.
Rajoriya, N., Combet, C., Zoulim, F., Janssen, H.L.A., 2017. How viral genetic variants Wang, G.H., Seeger, C., 1993. Novel mechanism for reverse transcription in hepatitis B
and genotypes influence disease and treatment outcome of chronic hepatitis B. Time viruses. J. Virol. 67, 6507–6512.
for an individualised approach? J. Hepatol. 67, 1281–1297. Wang, H., Kim, S., Ryu, W.S., 2009. DDX3 DEAD-Box RNA helicase inhibits hepatitis B
Rall, L.B., Standring, D.N., Laub, O., Rutter, W.J., 1983. Transcription of hepatitis B virus virus reverse transcription by incorporation into nucleocapsids. J. Virol. 83,
by RNA polymerase II. Mol. Cell Biol. 3, 1766–1773. 5815–5824.
Ramakrishnan, D., Xing, W., Beran, R.K., Chemuru, S., Rohrs, H., Niedziela-Majka, A., Wang, W.X., Li, M., Wu, X., Wang, Y., Li, Z.P., 1998. HNF1 is critical for the liver-specific
Marchand, B., Mehra, U., Zabransky, A., Dolezal, M., Hubalek, M., Pichova, I., function of HBV enhancer II. Res. Virol. 149, 99–108.
Gross, M.L., Kwon, H.J., Fletcher, S.P., 2019. Hepatitis B virus X protein function Wang, X., Hu, J., 2002. Distinct requirement for two stages of protein-primed initiation
requires zinc binding. J. Virol. 93. of reverse transcription in hepadnaviruses. J. Virol. 76, 5857–5865.
Reese, V.C., Oropeza, C.E., McLachlan, A., 2013. Independent activation of hepatitis B Watanabe, T., Sorensen, E.M., Naito, A., Schott, M., Kim, S., Ahlquist, P., 2007.
virus biosynthesis by retinoids, peroxisome proliferators, and bile acids. J. Virol. 87, Involvement of host cellular multivesicular body functions in hepatitis B virus
991–997. budding. Proc. Natl. Acad. Sci. U. S. A. 104, 10205–10210.
Ren, J.H., Hu, J.L., Cheng, S.T., Yu, H.B., Wong, V.K.W., Law, B.Y.K., Yang, Y.F., Xia, Y., Stadler, D., Lucifora, J., Reisinger, F., Webb, D., Hosel, M., Michler, T.,
Huang, Y., Liu, Y., Chen, W.X., Cai, X.F., Tang, H., Hu, Y., Zhang, W.L., Liu, X., Wisskirchen, K., Cheng, X., Zhang, K., Chou, W.M., Wettengel, J.M., Malo, A.,
Long, Q.X., Zhou, L., Tao, N.N., Zhou, H.Z., Yang, Q.X., Ren, F., He, L., Gong, R., Bohne, F., Hoffmann, D., Eyer, F., Thimme, R., Falk, C.S., Thasler, W.E.,
Huang, A.L., Chen, J., 2018. SIRT3 restricts hepatitis B virus transcription and Heikenwalder, M., Protzer, U., 2016. Interferon-gamma and tumor necrosis factor-
replication through epigenetic regulation of covalently closed circular DNA alpha produced by T cells reduce the HBV persistence form, cccDNA, without
involving suppressor of variegation 3-9 homolog 1 and SET domain containing 1A cytolysis. Gastroenterology 150, 194–205.
histone methyltransferases. Hepatology 68, 1260–1276. Xing, T., Zhu, J., Xian, J., Li, A., Wang, X., Wang, W., Zhang, Q., 2019. miRNA-548ah
Riviere, L., Gerossier, L., Ducroux, A., Dion, S., Deng, Q., Michel, M.L., Buendia, M.A., promotes the replication and expression of hepatitis B virus by targeting histone
Hantz, O., Neuveut, C., 2015. HBx relieves chromatin-mediated transcriptional deacetylase 4. Life Sci. 219, 199–208.
repression of hepatitis B viral cccDNA involving SETDB1 histone methyltransferase. Xu, L., Wu, Z., Tan, S., Wang, Z., Lin, Q., Li, X., Song, X., Liu, Y., Song, Y., Zhang, J.,
J. Hepatol. 63, 1093–1102. Peng, J., Gao, L., Gong, Y., Liang, X., Zuo, X., Ma, C., 2018. Tumor suppressor ZHX2
Riviere, L., Quioc-Salomon, B., Fallot, G., Halgand, B., Feray, C., Buendia, M.A., restricts hepatitis B virus replication via epigenetic and non-epigenetic manners.
Neuveut, C., 2019. Hepatitis B virus replicating in hepatocellular carcinoma encodes Antivir. Res. 153, 114–123.
HBx variants with preserved ability to antagonize restriction by Smc5/6. Antivir. Yamada, M., Oeda, A., Jung, J., Iijima, M., Yoshimoto, N., Niimi, T., Jeong, S.Y., Choi, E.
Res. 172, 104618. K., Tanizawa, K., Kuroda, S., 2012. Hepatitis B virus envelope L protein-derived bio-
Rost, M., Doring, T., Prange, R., 2008. gamma2-Adaptin, a ubiquitin-interacting adaptor, nanocapsules: mechanisms of cellular attachment and entry into human hepatic
is a substrate to coupled ubiquitination by the ubiquitin ligase Nedd4 and functions cells. J. Contr. Release 160, 322–329.
in the endosomal pathway. J. Biol. Chem. 283, 32119–32130. Yan, H., Zhong, G., Xu, G., He, W., Jing, Z., Gao, Z., Huang, Y., Qi, Y., Peng, B., Wang, H.,
Rost, M., Mann, S., Lambert, C., Doring, T., Thome, N., Prange, R., 2006. Gamma- Fu, L., Song, M., Chen, P., Gao, W., Ren, B., Sun, Y., Cai, T., Feng, X., Sui, J., Li, W.,
adaptin, a novel ubiquitin-interacting adaptor, and Nedd4 ubiquitin ligase control 2012. Sodium taurocholate cotransporting polypeptide is a functional receptor for
hepatitis B virus maturation. J. Biol. Chem. 281, 29297–29308. human hepatitis B and D virus. Elife 1, e00049.
Schulze, A., Gripon, P., Urban, S., 2007. Hepatitis B virus infection initiates with a large Yang, G., Feng, J., Liu, Y., Zhao, M., Yuan, Y., Yuan, H., Yun, H., Sun, M., Bu, Y., Liu, L.,
surface protein-dependent binding to heparan sulfate proteoglycans. Hepatology 46, Liu, Z., Niu, J.Q., Yin, M., Song, X., Miao, Z., Lin, Z., Zhang, X., 2019. HAT1 signaling
1759–1768. confers to assembly and epigenetic regulation of HBV cccDNA minichromosome.
Sekiba, K., Otsuka, M., Ohno, M., Yamagami, M., Kishikawa, T., Suzuki, T., Ishibashi, R., Theranostics 9, 7345–7358.
Seimiya, T., Tanaka, E., Koike, K., 2019. Inhibition of HBV transcription from Yao, Y., Yang, B., Cao, H., Zhao, K., Yuan, Y., Chen, Y., Zhang, Z., Wang, Y., Pei, R.,
cccDNA with nitazoxanide by targeting the HBx-DDB1 interaction. Cell Mol. Chen, J., Hu, X., Zhou, Y., Lu, M., Wu, C., Chen, X., 2018. RBM24 stabilizes hepatitis
Gastroenterol. Hepatol. 7, 297–312. B virus pregenomic RNA but inhibits core protein translation by targeting the
Sheraz, M., Cheng, J., Tang, L., Chang, J., Guo, J.T., 2019. Cellular DNA topoisomerases terminal redundancy sequence. Emerg. Microb. Infect. 7, 86.
are required for the synthesis of hepatitis B virus covalently closed circular DNA. Yao, Y., Yang, B., Chen, Y., Wang, H., Hu, X., Zhou, Y., Gao, X., Lu, M., Niu, J., Wen, Z.,
J. Virol. 93. Wu, C., Chen, X., 2019. RNA-binding motif protein 24 (RBM24) is involved in
Stahl, M., Beck, J., Nassal, M., 2007. Chaperones activate hepadnavirus reverse pregenomic RNA packaging by mediating interaction between hepatitis B virus
transcriptase by transiently exposing a C-proximal region in the terminal protein polymerase and the epsilon element. J. Virol. 93.
domain that contributes to epsilon RNA binding. J. Virol. 81, 13354–13364. Yuh, C.H., Ting, L.P., 1991. C/EBP-like proteins binding to the functional box-alpha and
Stieler, J.T., Prange, R., 2014. Involvement of ESCRT-II in hepatitis B virus box-beta of the second enhancer of hepatitis B virus. Mol. Cell Biol. 11, 5044–5052.
morphogenesis. PloS One 9, e91279. Zhang, P., McLachlan, A., 1994. Differentiation-specific transcriptional regulation of the
Sun, S., Nakashima, K., Ito, M., Li, Y., Chida, T., Takahashi, H., Watashi, K., Sawasaki, T., hepatitis B virus nucleocapsid gene in human hepatoma cell lines. Virology 202,
Wakita, T., Suzuki, T., 2017. Involvement of PUF60 in transcriptional and post- 430–440.
transcriptional regulation of hepatitis B virus pregenomic RNA expression. Sci. Rep.
7, 12874.

9
S. Tsukuda and K. Watashi Antiviral Research 182 (2020) 104925

Zhang, P., Raney, A.K., McLachlan, A., 1993. Characterization of functional Sp1 Zhou, L., Ren, J.H., Cheng, S.T., Xu, H.M., Chen, W.X., Chen, D.P., Wong, V.K.W., Law, B.
transcription factor binding sites in the hepatitis B virus nucleocapsid promoter. Y.K., Liu, Y., Cai, X.F., Tang, H., Yu, H.B., Hu, J.L., Hu, Y., Zhou, H.Z., Ren, F., He, L.,
J. Virol. 67, 1472–1481. Hu, Z.W., Jiang, H., Xu, H.Y., Huang, A.L., Chen, J., 2019. A functional variant in
Zhang, W., Chen, J., Wu, M., Zhang, X., Zhang, M., Yue, L., Li, Y., Liu, J., Li, B., Shen, F., ubiquitin conjugating enzyme E2 L3 contributes to hepatitis B virus infection and
Wang, Y., Bai, L., Protzer, U., Levrero, M., Yuan, Z., 2017. PRMT5 restricts hepatitis maintains covalently closed circular DNA stability by inducing degradation of
B virus replication through epigenetic repression of covalently closed circular DNA apolipoprotein B mRNA editing enzyme catalytic subunit 3A. Hepatology 69,
transcription and interference with pregenomic RNA encapsidation. Hepatology 66, 1885–1902.
398–415.

10

You might also like