Sie sind auf Seite 1von 12

3

Molecular Virology of Hepatitis E Virus


R. Prasida Holla, MSc1, Imran Ahmad, MSc1, Zulfazal Ahmad, PhD1 Shahid Jameel, PhD1

1 Virology Group, International Centre for Genetic Engineering and Address for correspondence Shahid Jameel, PhD, ICGEB, Aruna Asaf
Biotechnology (ICGEB), New Delhi, India, Ali Marg, New Delhi 110067, India (e-mail: shahid@icgeb.res.in).

Both authors contributed equally to the article.

Semin Liver Dis 2013;33:314.

Abstract Hepatitis E virus (HEV) is the causative agent of hepatitis E. It is a nonenveloped virus
Keywords with a 7.2 kilobases positive-stranded RNA genome. The molecular virology of HEV is
hepatitis E getting better understood with the development of replicons and in vitro infection
HEV systems, and the discovery of related viruses that infect animal species other than
ORF1 humans. This review focuses on the virology of HEV and updates the current knowledge
ORF2 on the HEV genome and its constituent proteinsORF1, ORF2, and ORF3, and the viral

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


ORF3 life cycle.
RNA replication

Hepatitis E virus (HEV) is the causative agent of hepatitis E, an nized as a species, each dominant in a given geographic area
acute viral hepatitis that is endemic to many resource-limited (Fig. 1). Genotype 1 includes strains from Asia and Africa;
regions of the world.1 According to the World Health Organi- genotype 2 includes a Mexican strain and few variants from
zation, there are approximately 20 million incident HEV Africa; genotypes 3 and 4 include human and swine HEV
infections every year, with over 3 million acute cases of strains from industrialized countries and Asia (particularly
hepatitis E and 70,000 hepatitis Erelated deaths.2 Most China), respectively.4,5 While genotypes 1 and 2 are restricted
of these are in high-prevalence areas in East and South Asia to humans, genotypes 3 and 4 have a broader host range and
where sanitary conditions are poor and there is frequent are zoonotic. The human and swine HEV strains show exten-
contamination of drinking water with fecal matter. Large sive serological cross reactivity with a single serotype.
outbreaks of hepatitis E have also been reported from these Studies on HEV transmission and pathogenesis as well as
areas, as from other areas of overcrowding and poor sanita- preclinical vaccine development studies were performed in
tion such as refugee camps. nonhuman primates such as cynomolgus, rhesus and owl
Hepatitis E was recognized to be caused by a distinct agent monkeys, and chimpanzees.68 Pigs have also been used for
in the 1970s from outbreak investigations undertaken by transmission and molecular studies.9 But a small animal
Khuroo and colleagues in the Kashmir, India, and by Purcell model for human HEV is still elusive, which has hampered
and colleagues who retrospectively studied the 19551956 virological studies. Cell culture systems based on replicon
outbreak of jaundice in New Delhi, India. The historical RNA transfection and those using the human, swine, and
aspects of hepatitis E and HEV are covered in greater detail rabbit viruses have recently become available, which are
in a recent review.3 Viruses similar to HEV have been discov- discussed below. These are likely to result in a more complete
ered from pigs (swine HEV) and chickens (avian HEV), and understanding of hepatitis E virology.
more recently from bats, rats, rabbits, and sh. The successful
transmission of swine HEV to macaques, which are a model
Hepatitis E Outbreaks
for human HEV transmission, and reports showing nucleotide
sequence identity between viruses recovered from uncooked Many outbreaks of hepatitis E have been recorded from
meat and humans who became infected after consuming the endemic areas. The rst recorded outbreak in Delhi, India
meat, strongly support HEV as a zoonosis. in 1955 to 1956 with an estimated 29,000 cases was retro-
HEV is a nonenveloped virus, which is 27 to 34 nm in size spectively conrmed about two decades later when stored
and is classied as a Hepevirus in the family Hepeviridae.4 sera were tested negative for hepatitis A and B10; this was also
Within this genus at least four genotypes of HEV are recog- the case with a subsequent outbreak from Ahmedabad, India

Issue Theme Hepatitis E in 2013: Copyright 2013 by Thieme Medical DOI http://dx.doi.org/
Essential Facts, Emerging Concepts Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0033-1338110.
and Challenges; Guest Editor, Kris New York, NY 10001, USA. ISSN 0272-8087.
Krawczynski, MD, PhD Tel: +1(212) 584-4662.
4 HEV Virology Holla et al

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


Fig. 1 Global distribution of hepatitis E virus (HEV). The world map shows an updated distribution pattern of HEV genotypes among infected
humans as well as the distribution of swine HEV genotypes in infected pig populations.

in 1975 to 1976. The proposal that enteric non-A, non-B reading frames (ORFs) called orf1, orf2, and orf3, the viral
hepatitis (ET-NANBH) was due to a distinct agent was made genome also has short 5- and 3-untranslated regions (UTRs),
following the investigation of a 1978 outbreak in Kashmir, a conserved 58-nucleotide region within orf1 and a sequence
India.11 The viral inoculum was rst passaged into nonhuman closer to the 3 end of orf1, which is homologous to the
primates and into a human volunteer from a 1983 outbreak junction region found in alphaviruses (Fig. 2).18 These
among Soviet troops stationed in Afghanistan.12 Subsequent- structures are proposed to be important for HEV RNA repli-
ly, two small outbreaks affecting approximately 200 people cation.19 The region between the end of orf1 and start of orf3/
were reported in 1986 to 1987 from Mexico.13 A prolonged orf2 is complex and also contains regulatory elements.20
epidemic from late 1986 to 1988 was reported from the The cloning and sequencing of the HEV genome was rst
Xinjiang Uyghur autonomous region in western China14; a reported from cDNA libraries made from the bile of macaques
subsequent outbreak was reported from Nepal in 1988. In experimentally infected with stool samples from hepatitis E
1991, a large epidemic occurred in Kanpur, India in which patients.18,21 Full-length genomic sequences from various
79,000 people were affected15; a massive outbreak in China in genotypes and geographic isolates of HEV have since become
the same year affected more than 100,000 people. Nepal saw available.2224 Three RNA species of 7.2, 3.7, and 2 kb,
the recurrence of a hepatitis E outbreak in 1994, but the strain designated as the genomic and two subgenomic RNAs, re-
responsible was different than the 1988 outbreak. Several spectively, were detected in the liver tissue of macaques
other outbreaks have been reported from Asia and Africa. The experimentally infected with HEV.18 In the original model,
Uganda outbreak of 2007 had almost 11,000 cases and 160 while the 7.2 kb RNA was proposed to be translated into the
fatalities.16 In 2004, the refugee camps in Sudan and Chad ORF1 protein, the 3.7 and 2 kb subgenomic RNAs translated
experienced hepatitis E, which were caused by viruses ge- into the ORF3 and ORF2 proteins, respectively.18 However,
netically related to Asian outbreaks, but some isolates were other workers were unable to nd the 3.7 kb RNA in animal
closer to the Mexican genotype 2 virus.17 Most recently, an livers or in cell culture models of HEV replication. In stable
outbreak was reported from Bangladesh in June 2011 and Huh-7 cell lines made from functional HEV RNA replicons, the
another in June 2012 from a village in Maharashtra, India, genomic RNA and only one subgenomic RNA of 2.2 kb were
which had more than 3000 cases and 18 fatalities. A common observed.25 The capped subgenomic RNA initiated at nucleo-
feature of all hepatitis E outbreaks was contamination of tide position 5122, which was downstream of the rst two
drinking water supplies with sewage or fecal material. methionine codons in orf3, and was bicistronic for the
translation of ORF3 and ORF2 (Fig. 2). This model rational-
ized the reading frame differences observed in genotype 4
The HEV Genome
HEV isolates and was further conrmed by intrahepatic
The genome of HEV is a single-stranded positive-sense RNA of inoculation of wild-type and mutant genotype 3 swine HEV
 7.2 kb, which has features typical of a eukaryotic mRNA, replicons into pigs26 and from PLC/PRF/5 hepatic cells that
including a 7-methylguanine cap at its 5 end and a poly(A) were either transfected with an infectious genotype 3 RNA or
stretch at the 3 end. Besides three partially overlapping open infected with HEV genotype 4.27 In both cases, only the 2.2 kb

Seminars in Liver Disease Vol. 33 No. 1/2013


HEV Virology Holla et al 5

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


Fig. 2 The hepatitis E virus (HEV) genome. The  7.2 kb positive sense RNA genome of HEV has a 7-Me-G cap at its 5 end and a poly A tail at its 3
end. There are short stretches of untranslated regions (UTR) at the 5 and 3 ends. The three open reading frames (ORFs) are shown. The orf1
(nucleotides 285109) encodes a nonstructural polyprotein that contains the methyltransferase (MeT), Y, papain-like cysteine protease (PCP),
variable (V), X(macro), RNA helicase (Hel), and RNA-dependent RNA polymerase (RdRp) domains. The orf2 (nucleotides 51477129) and orf3
(nucleotides 51075477) are translated from a 2.2 kb subgenomic RNA generated during viral replication. The ORF2 protein has three N-linked
glycosylation sites (red dots) at residues 137, 310, and 547. The domain structure of the ORF3 protein and a phosphorylation site (red diamond) at
residue Ser-71 are shown.

subgenomic RNA species, whose 5 end mapped to nucleotide other sites in China, with the strains clustered into three
5122, was observed. A recent review provides more details on distinct groups within HEV genotype 3.34 Finally, a virus that
HEV RNA species, regulatory elements on the viral genome, infects various species of trout sh was isolated from the
and their role in genome replication.20 cutthroat trout; it bears only 18 to 27% sequence similarity to
avian or mammalian HEVs and may be classied as a new
Genetic Variants genus within the family Hepeviridae.35
Comparative analysis of the genomic sequences of multiple It is suspected that a high level of genomic intermixing
HEV isolates has revealed extensive genomic diversity and the takes place in animals and in patients. Recombination in HEV
identication of four major genotypes and several subtypes genomes was recently reported.36 Phylogenetic and recom-
within each genotype.28 A virus with a 6.6 kb RNA genome bination analyses of complete HEV genomes showed three
and 50% nucleotide sequence identity with mammalian potentially signicant intra- and intergenotype recombina-
HEVs, called avian HEV, was isolated from chickens affected tion events. Compartmentalization of HEV quasispecies be-
by the hepatitis-splenomegaly syndrome.29,30 Though ini- tween serum and cerebrospinal uid (CSF) was observed in a
tially considered as HEV genotype 5, avian HEV is now kidney transplant patient with chronic hepatitis E.37 The
proposed as a new species within the family Hepeviridae.4 genetic variability among quasispecies from solid-organ
Recently, HEV-like viruses were also isolated and character- transplant patients who developed hepatitis E was found to
ized from Norway rats31 and ferrets.32 The rat HEV has a be lower in those who cleared the virus as compared with
sequence identity of 55 to 59% with HEV genotypes 1 to 4, those who progressed to chronicity.38
and ferret HEV has an identity of 72.3% to rat HEV. A unique Chronic hepatitis E infection in immunocompromised
strain of HEV was identied from farmed rabbits in China, individuals has revealed the presence of viral strains carrying
which shared 74 to 79% nucleotide sequence identity to insertions from human genes. These recombinants were
existing HEV strains, and 46% identity to avian HEV.33 High barely detectable in feces from patients, but were selected
rates of HEV seropositivity were recently reported in rabbits with serial passage on cells in culture. The Kernow-C1 strain
raised on farms in Inner Mongolia, China.34 Sequence analysis of HEV was isolated from the feces of a chronically infected
showed 89.3 to 95.9% identity to rabbit HEV strains from HIV-positive patient, and was subsequently passaged on

Seminars in Liver Disease Vol. 33 No. 1/2013


6 HEV Virology Holla et al

HepG2/C3A cells.39 After six passages, the isolated viral capped, it was expected that the HEV genome would encode a
species were found to carry insertions from the host genome, methyltransferase. The capping of mRNA is well character-
which included 174 nucleotides or 117 nucleotides from the ized in mammalian, yeast, and several viral systems, in which
S17 or S19 ribosomal protein genes, respectively, or 114 it follows a common mechanism, which involves RNA tri-
nucleotides from a GTPase gene; point mutations were also phosphatase, guanyltransferase, and guanine-7-methyltrans-
observed in the hypervariable region (HVR) within orf1.39 The ferase activities.47 A 110-kDa protein that included amino
S17 insertion and point mutations conferred an adaptive acids 1 to 979 of ORF1 was shown to contain the guanyl-
advantage in culture, and expanded the host range and transferase and guanine-7-methyltransferase activities.48
tropism of the virus.39 The S17 fragment from the Kernow The RNA triphosphatase activity was suggested to be part
virus was also cloned into an infectious clone of the HEV of the HEV helicase protein.49
genotype 1 Sar-55 strain. Although the virus produced from Two reports have shown the presence of a 7-methylgua-
this clone did not show any adaptive advantage in HepG2/C3A nine cap on HEV genomic RNA, and its requirement for
cells, it had a broader tropism and infected a hamster cell infectivity.24,50 This cap structure is required for efcient
line.40 The reasons for better growth in culture and expanded binding of mRNAs to the ribosome, as well as for viral RNAs to
tropism are unclear at this time. evade activation of interferon-mediated innate immunity.
The 5 triphosphate (5ppp) group on an uncapped RNA
molecule is a potent activator of the interferon response.51
HEV Proteins
The HEV genome encodes three proteins, which are called the Protease

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


ORF1, ORF2, and ORF3 proteins. Although the kinetics of A papain-like cysteine protease (PCP) was also predicted
expression of these proteins during the viral life cycle is not within the ORF1 polyprotein.43 This prediction was strength-
fully understood, antibodies to these proteins in infected ened by conservation of the X domain in HEV, which in other
humans and experimental animals is a conrmation of their systems was found exclusively in association with the PCP.
expression during HEV infection.41,42 Although this alignment placed the putative PCP domain in
the region of amino acids 433 to 592 of the ORF1 polyprotein,
The ORF1 Protein no functional activity for the HEV protease has so far been
The ORF1 nonstructural polyprotein of HEV is 1693 amino described. This was further conrmed recently based on
acids (180 kilodaltons [kDa]) and contains several function- biochemical data and the lack of conservation of the catalytic
al motifs and domains present in the nonstructural proteins Cys-His dyad in a large number of HEV isolates.52 However, a
of other positive-stranded RNA viruses.43 These include the deubiquitination activity was recently reported in a Met-PCP
methyltransferase (MeT), papain-like cysteine protease fusion protein, and is proposed to play a role in combating the
(PCP), RNA helicase (Hel) and RNA-dependent RNA polymer- cellular antiviral response.53 Signicantly, the cellular ubiq-
ase (RdRp) (Fig. 2). Some uncharacterized domains homol- uitin-proteosome system and the free ubiquitin pool also
ogous to other animal and plant positive-strand RNA viruses appear to be important for HEV replication.54
are also found within the ORF1 protein. These include the Y
domain, the X domain (now called the macro domain), and a RNA Helicase
polyproline or hypervariable region that shows high levels of Many positive-stranded RNA viruses encode a RNA helicase,
nucleotide (and amino acid) variation between HEV isolates which is essential for replication of the viral genome of these
(Fig. 2). A bioinformatic analysis of the HEV ORF1 protein43 viruses. The putative RNA helicase of HEV was shown to
proposed many putative biochemical activities, some of contain all the conserved motifs found in SF-1 type helicases
which have now been characterized in greater detail. It is and to have the nucleotide triphosphatase (NTPase) as well as
not entirely clear whether the ORF1 polyprotein is processed RNA-binding domains.43 This was biochemically conrmed
into biochemically distinct units, as is the case with many recently for a region of the ORF1 protein encompassing amino
positive-strand RNA viruses. Expression of the ORF1 poly- acids 960 to 1204.55 The enzyme hydrolyzed all rNTPs, as also
protein by itself showed some processing when expressed in dNTPs albeit with a lower efciency, and showed unwinding
mammalian cells using recombinant vaccinia viruses44 or in activity only on RNA duplexes with 5 overhangs. The HEV
insect cells using recombinant baculoviruses,45 but not when helicase belongs to the 5!3 class of SF-1 family of helicases
expressed in Escherichia coli or HepG2 hepatoma cells.46 that are also found in other positive-strand RNA viruses such
However, processing was reported in HepG2 cells transfected as alphaviruses, arteriviruses, coronaviruses, and rubiviruses.
with a genomic replicon of HEV.23 No viral infection study has
so far addressed expression and processing of the ORF1- RNA-Dependent RNA Polymerase
derived nonstructural polyprotein. The RdRp replicates the genomic RNA through an antige-
nomic RNA intermediate, and is an essential enzyme encoded
Methyltransferase by all RNA viruses. A strong homology of the HEV ORF1 region
An alignment of the ORF1 polyprotein suggested that amino encompassing amino acids 1207 to 1693 was observed with
acids 60 to 240 might serve as the viral methyltransferase.43 the RdRp proteins of other positive-strand RNA viruses.43 The
Viral methyltransferases catalyze the capping of viral RNA, RdRp region comprising nucleotides 3546 to 5106 of HEV was
and because the HEV genomic and subgenomic RNAs are expressed in E. coli and the puried recombinant protein

Seminars in Liver Disease Vol. 33 No. 1/2013


HEV Virology Holla et al 7

bound the 3 end of HEV genomic RNA.56 The recombinant RNA genome.19 When expressed in animal cells in culture,
RdRp also used the 3 end of HEV RNA as a template to nonglycosylated and glycosylated ORF2 proteins of 74 kDa
synthesize the complementary strand in vitro. A  40 kDa C- and 88 kDa were observed.65 The nascent protein contains a
terminal region of the ORF1 polyprotein, expressed as a N-terminal signal sequence that translocates it into the
fusion with the enhanced green uorescent protein, localized endoplasmic reticulum, where it acquires N-linked glycosyl-
to the endoplasmic reticulum (ER) and was able to copy HEV ation65 at conserved asparagine (Asn) residues 137, 310, and
plus-strand RNA transfected into these cells.57 These studies 562 (Fig. 2).66 This and the requirement of glycosylation
also suggest that analogous to several other RNA viruses, sites on the ORF2 protein for the formation of infectious virus
intracellular (ER) membranes are the likely sites for HEV particles was subsequently shown using infectious repli-
replicase localization and possibly RNA replication as well. cons67 and HEV that propagates efciently in cultured PLC/
PRF/5 cells.68 A genotype 3 ORF2 protein expressed using
Macro Domain recombinant vaccinia viruses was also glycosylated and lo-
Macro domains have homology to the nonhistone domain of calized to the ER, Golgi, and surface of infected cells.69
the histone macroH2A and are found in a variety of bacterial, A N-terminal truncated ORF2 protein (aa 112660) of 58
archaeal, and eukaryotic proteins, where they contribute to kDa and a 50 kDa form were efciently secreted as virus-like
ADP-ribose metabolism and posttranslational modica- particles (VLPs) of 23 to 24 nm in the culture medium of
tions.58 These are also expressed in members of a few viral insect cells infected with recombinant baculoviruses.70 When
families in which viral RNA is replicated in the cytoplasm in expressed similarly, the full-length ORF2 protein was proc-
membrane-associated replication complexes; these include essed into 63 kDa, 56 kDa, and 53 kDa proteins, which

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


all members of Coronaviridae, rubella virus, and alphaviruses correspond to truncated proteins spanning amino acids 112
(Togaviridae).59 Macro domain proteins hydrolyze ADP-ri- to 660, 112 to 607, and 112 to 578, respectively.71 Cryoelec-
bose 1-phosphate (ADPR-1P), a product of cellular pre-tRNA tron microscopy and image reconstruction methods showed
splicing, and associate with poly(ADP-ribose) polymerase-1 these VLPs to contain protruding dimers at the icosahedral
(PARP-1),60 suggesting some role in cellular apoptosis. Two twofold symmetry axes such that the 60 monomers were
reports describe the macro (erstwhile X) domain within organized as 30 morphological units.72 Subsequently, amino
amino acids 775 to 960 of the HEV ORF1 polyprotein.61,62 It acids 126 to 601 in the ORF2 protein were found to be
is suggested that viral macro domains function as poly(ADP- essential for the formation of secreted VLPs in this expression
ribose)-binding modules and may have a role in viral RNA system.73 X-ray crystal structures show the ORF2 protein
replication and/or transcription.61 Because some of the mac- monomers to contain three distinct domains called the shell
ro domains, including that of HEV, can also bind poly(ADP- (S), middle (M), and protruding (P) domains.74,75 These and
ribose) in the presence of poly(A), they could recruit poly other structural details are covered in a recent review.76
(ADP-ribose)-modied cellular factors to the replication com- Recombinant ORF2 proteins expressed in E. coli can also
plex while bound to viral polyadenylated RNA. assemble into higher-order structures, as exemplied by the
p239 (aa 368606), E2 (aa 394606) and E2a (459660)
Polyproline Region proteins that predominantly occur as homodimers and that
The polyproline region (PPR) or hypervariable region in the model the dominant antigenic and neutralizing determinants
ORF1 polyprotein is classied as an intrinsically disordered on the HEV surface.77,78 The crystal structure of the E2s
region (IDR), rich in polar and charged amino acids. A recent protein (aa 455602) shows it to form a tight homodimer
study63 shows the PPR mutation rate to be similar to the rest that is essential for HEV interaction with the host cell, and
of orf1, but due to a higher substitution rate within the PPR, contains the HEV neutralizing antibody recognition site.79
there is greater promiscuity in the rst and second codons. Various studies have characterized the neutralizing do-
Due to a preference for cytosine (C) in the rst and second mains of the ORF2 capsid protein through phage display
positions, there is increased frequency of proline residues in methods80 or with multiple overlapping peptides and trun-
this region. Sequence conservation and lower rates of substi- cated ORF2 proteins.81 The minimal neutralizing domain was
tution in the PPR in genotype 1 compared with genotypes 3 mapped to residues 458 to 607.82 When combined with
and 4 may suggest an increased need for adapting to a wider structural details, these mapping studies show the P domain
host range by zoonotic strains. In another recent study, a of the ORF2 protein to contain its neutralization epitope.74,75
three-dimensional model of the PPR region was used for Immunological and structural studies on the ORF2 protein
function prediction using gene ontology annotations.64 Sev- have also provided a basis for hepatitis E vaccine develop-
eral putative functions were found, such as peptide cleavage ment. Two recombinant hepatitis E vaccines based on recom-
sites, sites modied by enzymes, and sites that bind to binant ORF2 proteins expressed as VLPs in insect cells83 or in
proteins, nucleotides, and metal ions, indicating a potential E. coli84 have undergone successful clinical testing in humans.
role in viral replication. These proposals await experimental The ORF2 particles have also been used as an epitope presen-
validation. tation system,85 which when combined with the ability of
these VLPs to render successful oral immunization,86 can
The ORF2 Protein provide a powerful vaccine platform. A recent study com-
The ORF2 protein is the viral capsid protein of 660 amino pared the cross-protective ability of N-terminal truncated
acids (Fig. 2), which was proposed to encapsidate the viral ORF2 proteins from swine, rat, and avian HEV in a pig

Seminars in Liver Disease Vol. 33 No. 1/2013


8 HEV Virology Holla et al

challenge model infected with genotype 3 human HEV. mentous intracellular distribution, attributed to its
Although a strong IgG anti-HEV response was observed, there localization to endosomes98 and its interaction with micro-
was only partial cross protection.87 This has important im- tubules,99 respectively.
plications for current and future vaccines, especially against The D1 domain is very cysteine-rich and is required for the
novel zoonotic strains of HEV. ORF3 protein to associate with the cytoskeleton,97 to bind
Recombinant ORF2 capsid proteins have also been used to microtubules99 and a MAPK phosphatase.100 The P1 domain
model the interactions of HEV with its target cells. The contains a PMSP motif in which Ser-71 is phosphorylated by
puried p239 protein, which antigenically and structurally MAPK, in transfected cells as well as in vitro97; whether the
mimics the HEV capsomere can bind and penetrate cells ORF3 protein expressed during viral infection is also phos-
susceptible to HEV infection in vitro.88 In interaction studies, phorylated has not been tested. The P2 domain has two
p239 bound to cellular Grp78/BiP, -tubulin and heat shock overlapping PXXP motifs, which have been described in
protein 90 (Hsp90), with Hsp90 binding being important for many viral and cellular signal-transducing proteins and
its intracellular trafcking.89 The ORF2 VLPs bound Huh7 bind the Src homology 3 (SH3) domains found in other
liver cells in an interaction dependent upon heparan sulfate proteins. This domain was shown to be responsible for
proteoglycans (HSPGs) present on the cell surface.90 Our interaction of the ORF3 protein with many cellular SH3
studies also show the p239 VLPs to bind cell lines susceptible domain-containing proteins.101
to HEV infection; these are being used to identify the cellular The ability of the ORF3 protein to interact with multiple
receptor for HEV (R.P. Holla, Z. Ahmad, and S. Jameel, unpub- cellular proteins suggested its potential role in optimizing the
lished results). cellular environment for viral infection and replication.

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


For nonenveloped viruses, the interactions of their capsid Though most of the studies are based on ORF3 overexpression
proteins with cellular proteins are important for virus entry, in hepatic cell lines, they provide insights into the versatile
intracellular trafcking, and signaling,91 as well as for capsid nature of this protein. The ORF3 protein activates the extra-
assembly and virus egress. When expressed in animal cells, cellularly regulated kinase (Erk), a member of the MAPK
the ORF2 protein localizes to the ER, where it causes ER stress family,101 and this depends upon its ability to bind and
and some of the protein is retrotranslocated to the cytoplasm inactivate an Erk-specic MAPK phosphatase.100 Cells ex-
through an ER-associated degradation (ERAD) pathway.92 A pressing the ORF3 protein also displayed higher levels of
recent study shows ORF2-dependent phosphorylation of the hexokinase and oligomeric forms of the voltage-dependent
eukaryotic initiation factor 2a (eIF2a), increased expression of anion channel (VDAC) protein, which lead to attenuation of
the ATF-4 transcription factor, and activation of the CHOP mitochondrial death signaling.102 Subcellular localization of
promoter.93 Even though this ER stress pathway leads to the ORF3 protein to early and recycling endosomes, and its
apoptosis, none was observed in ORF2-expressing cells, pos- interaction with CIN85, a multidomain adaptor protein im-
sibly due to its interaction with heat shock protein 72 plicated in the downregulation of receptor tyrosine kinases,
(Hsp72), which triggers an anti-apoptotic response.93 The delays postinternalization trafcking of the activated growth
glycosylated form of the ORF2 protein also inhibited the factor receptors.98,103 The activation of Erk and effects on
nuclear factor kappa B (NFB) pathway by reducing the growth factor receptors are proposed to promote cell survival
proteasome-mediated degradation of inhibitor of kappa B and proliferation. Another consequence of its effects on
(IB), which was due to ORF2 protein interaction with -TRCP, trafcking is reduced levels of phospho-STAT3 (pSTAT3) in
a component of the ubiquitination complex.94 Whether these the nuclei of ORF3-expressing cells.98 This results in de-
interactions modulate HEV entry, intracellular trafcking, creased expression of acute-phase response proteins and
assembly, or egress remains to be seen in an infection model. attenuation of a major inammatory pathway in the liver,
which would again create an environment supportive of viral
The ORF3 Protein replication. Reduced levels of acute phase proteins are also
The ORF3 protein is 114 amino acids long and is dispensable observed in the plasma of hepatitis E patients.104
for replication of HEV in vitro in cell lines,95 but is required for Various other ORF3-interacting cellular proteins have been
infection in the macaque model of experimental infection.96 identied through interaction screens. These include the -1-
This suggests that the ORF3 protein functions as a viral microglobulin and bikunin precursor protein (AMBP) and its
accessory protein, and is likely to affect the host response constituent -1-microglobulin and bikunin, brinogen
to infection. Primary sequence analysis of the ORF3 protein chain, and hemopexin.105108 Increased secretion of -1-
did not show homology to any other protein. It contains two microglobulin was observed from ORF3-expressing cells,
large N-terminal hydrophobic domains, D1 (aa 723) and D2 mediated by the tumor susceptibility gene 101 (Tsg101)
(aa 2853), and two proline-rich domains, P1 (aa 6677) and protein, a member of the endosomal-sorting complex, which
P2 (aa 95111) (Fig. 2). In mammalian cells, the 13 kDa bound the ORF3 protein through a conserved PSAP motif in its
protein was phosphorylated at a single serine residue within P2 domain.109 Because -1-microglobulin is immunosup-
the P1 domain by the cellular mitogen-activated protein pressive, this was proposed to protect virus-infected cells
kinase (MAPK)97; this was also the case in cells transfected in the liver.109 The urine of hepatitis E patients also showed
with the HEV genomic RNA.23,95,96 Subcellular fractionation increased -1- microglobulin levels compared with hepatitis
showed the ORF3 protein to associate with the cytoskele- B patients and healthy controls.104 Hemopexin is a heme-
ton,97 and confocal microscopy revealed punctate and la- binding acute-phase plasma protein, which protects cells

Seminars in Liver Disease Vol. 33 No. 1/2013


HEV Virology Holla et al 9

from hemoglobin-mediated oxidative damage and together to virus budding.117 Viral late domains interact with mem-
with transferrin and haptoglobin plays an important role in bers of the VPS pathway, and redirect the complexes to the
iron homeostasis. The ORF3 protein binds hemopexin site of virus budding on the plasma membrane.117 Although it
through its D2 domain, and this is proposed to aid in viral is a nonenveloped virus, the association of HEV with lipids,
infection.107 Reduced levels of hemopexin and haptoglobin endosomal localization of the ORF3 protein, and the presence
have also been observed in the plasma of hepatitis E pa- of a PSAP motif that is required for viral egress, suggests that
tients.104 The ORF3 protein also increased the expression of HEV follows the VPS pathway for its release from infected cells
many glycolytic enzymes through Erk-mediated stabilization (Fig. 3).
of the hypoxia inducible factor-1 (HIF-1) transcription fac-
tor.110 Finally, it modulates the phosphorylation and the
HEV Replication and In Vitro Propagation
nuclear localization of hepatocyte nuclear factor 4 (HNF4),
and through it the expression of many cellular genes that are The HEV replication cycle is poorly understood due to its
regulated by this liver-specic transcription factor.111 These inability to efciently propagate in vitro in cultured cells or to
studies support a prosurvival (or antiapoptotic) role for the infect small animal models. The original view of the HEV life
ORF3 protein, as also its effects on cellular energy homeosta- cycle118 was based on its genome analysis and analogy to
sis, innate host responses and transcription of cellular genes, other positive-strand RNA viruses; it is still generally rele-
which are likely to support HEV pathogenesis in the infected vant. However, in recent times in vitro expression and repli-
host. con studies have added some interesting details. An updated
Some recent studies have shown the ORF3 protein to be model of the HEV life cycle is shown in Fig. 3.

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


associated with virions and have proposed a role in virus Various transformed cell lines, such as the hepatic cell lines
egress. An ORF3-decient infectious cDNA clone replicated Huh7, PLC/PRF/5 and HepG2, the lung carcinoma cell line
efciently in PLC/PRF/5 and A549 cells, but was defective in A549, and the colon carcinoma cell line Caco-2, have been
virus release.68 In an immunocapture PCR assay, the ORF3 shown to support HEV infection and replication. The binding
protein was found on the surface of cell culture generated and entry of HEV is poorly understood as no cellular receptor
HEV, which also showed a lower density than the ORF3- for the virus has been identied. The HSPGs present on cell
decient virus. These observations suggested the ORF3 pro- surface Syndecan type proteins are required as attachment
tein to be present on the virion surface in association with factors, but their role is largely to concentrate the virus on the
lipids, and to be important for HEV egress.68 Anti-ORF3 cell surface before its binding to and entry through a specic
antibodies were also able to capture HEV from the serum, receptor (Fig. 3, steps 1, 2).90 One recent report suggests
but not the feces of hepatitis E patients, the former banding at that HEV enters liver cells through receptor-dependent
a lower density compared with the latter.112 The HEV in clathrin-mediated endocytosis (Fig. 3, step 3).119 However,
serum appears to be associated with lipids, but this coat is alternate entry pathways are also possible (R.P. Holla and S.
possibly shed during its passage through the gut. Jameel, unpublished results). Following entry, Hsp90 and
The egress of a genotype 1 HEV from Caco-2 (intestinal) tubulin appear to be involved in intracellular trafcking,89
and Huh-7 (hepatoma) cells depended upon the expression of but little else is known about this process. The location and
a functional ORF3 protein.113 This protein accumulated in mechanism of uncoating to release viral RNA (Fig. 3, step 4)
polarized Caco-2 cells at the apical surface from which the is not known.
virus was released.113 In this as well as another study,114 a Once it is released in the cytosol, the viral RNA is directly
PSAP motif within the P2 domain of the ORF3 protein was translated into the ORF1 polyprotein (Fig. 3, step 5). The
found to be important for virus egress, but not for its viral methyltransferase, RNA triphosphatase, helicase and
accumulation at the apical end. One of the mutant viruses replicase activities are used to replicate the genomic RNA
showed no infection following intrahepatic inoculation in into negative-sense RNA intermediates (Fig. 3, step 6),
macaques, but another was infectious and the recovered virus which have been detected in replicon-transfected cells23 as
was found to have completely reverted to a wild-type ORF3 well as in the livers of experimentally infected macaques120
sequence. These results support previous observations that and pigs.9 The negative-strand RNA serves as a template for
the ORF3 requirement for infection.96 In this study also, the the synthesis of genomic as well as subgenomic positive-
ORF3 protein, in association with lipids, was found to coat the sense RNAs (Fig. 3, steps 7, 8), of which the latter are
virion surface. translated into the ORF2 and ORF3 proteins (Fig. 3, step
Recent studies show the requirement of an intact PSAP 9).2527 The ORF2 protein packages the genomic positive-
motif for the formation of membrane associated HEV par- sense RNA into progeny virions (Fig. 3, step 10). Recent
ticles with the ORF3 protein on their surface,114 and this to be evidence suggests that the ORF3 protein together with lipids
mediated by the cellular Tsg101 protein115; this holds true for coats this particle, possibly during the budding process
avian HEV as well.116 Several RNA viruses bear proline-rich (Fig. 3, step 11).
sequences in their late domains, which are required for viral Although hepatocytes are the most likely cell type in which
budding. These include P(S/T)AP and PPXY (X, any amino acid) HEV replicates, in vitro results also support infection and
motifs, which hijack host proteins in the vacuolar protein replication in nonhepatic cell types such as A549 lung carci-
sorting (VPS) pathway. This pathway gives rise to multi- noma cells and in Caco-2 colon carcinoma cells. In pigs
vesicular bodies (MVBs), which are topologically identical experimentally infected with swine HEV, negative-sense

Seminars in Liver Disease Vol. 33 No. 1/2013


10 HEV Virology Holla et al

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


Fig. 3 The hepatitis E virus (HEV) replication cycle. The viral particles are concentrated on the surface of target cells through heparin sulfate
proteoglycans acting as attachment factors (1) and subsequently bind a specic yet uncharacterized receptor (2), following which the particles are
internalized by endocytosis (3). The virus uncoats (4) to release genomic RNA that is translated in the cytoplasm into nonstructural proteins (5).
These nonstructural proteins include the RNA-dependent RNA polymerase that replicates the positive sense genomic RNA into negative sense
transcripts (6); the latter then act as templates for the synthesis of a 2.2 kb subgenomic RNA (7) as well as full-length positive sense transcripts (8).
The positive sense subgenomic RNA is translated into ORF2 (gray triangles) and ORF3 (blue circles) proteins (9). The ORF2 protein goes through
the endoplasmic reticulum and packages the genomic RNA to assemble new virions (10). The ORF3 protein is also associated with
endomembranes (11) and may aid in viral egress (12,13). Recent studies suggest that mature virions are associated with the ORF3 protein and
lipids (13), which are subsequently removed through a process that is not understood at present, to resume a fresh infection cycle.

replicative RNA intermediates were detected in the liver, but plant patients with chronic HEV infection,125127 severe
also in the small intestine, lymph node, and colon.121 In acute hepatitis E,128 and acute on chronic liver failure.129
hepatitis E patients, HEV RNA, but no viral replication, was
also detected in peripheral blood mononuclear cells.122
Summary
Since its discovery in the early 1970s, molecular biological
Antiviral Targets in HEV
tools have contributed signicantly toward understanding
As in many viruses, multiple steps in the HEV life cycle can the virology of HEV. Although the lack of efcient in vitro
also be potential targets for antiviral drug development. The culture systems and small animal models of viral infection
methyltransferase and guanyltransferase activities in the continue to be problems, recent work is beginning to over-
ORF1 protein being strictly virus-specic are good targets come some of these hurdles. Much of the early information
for antiviral development.48 The HEV RNA helicase is bio- and models of the HEV life cycle and genome replication were
chemically characterized and is essential for viral RNA repli- based on comparative information from better-characterized
cation.54 The determination of its structure is important to positive-strand RNA viruses. Viral ORFs have been expressed
understand how distinct it is from human helicases before in various heterologous systems and the proteins have been
being promoted as a good drug target. The HEV RdRp would characterized for their structure and function. With the
also be a potential target. However, only its binding to the recent availability of replicon- and infection-based in vitro
viral RNA genome,56 but not its biochemical activity, have so cell culture models, the molecular virology of HEV is becom-
far not been characterized. Because the RdRp enzyme is ing increasingly clear. This review summarizes the current
unique to RNA viruses and is absent in vertebrate hosts, knowledge on HEV virology.
panviral inhibitors targeting positive- or negative-strand
RNA viruses should be explored against this enzyme.
Interference with HEV RNA replication has been at- Acknowledgments
tempted using ribozymes and small interfering RNAs.123,124 The HEV work from the authors laboratory has been
Although such approaches are feasible in vitro, the delivery funded by grants from the Wellcome Trust, UK, National
and targeting of such inhibitors in vivo would be the real Institutes of Health, USA, Department of Biotechnology,
challenge. Several recent reports show the efcacy of Ribavi- India, and from ICGEB internal resources. All are gratefully
rin monotherapy in treating immunocompromised trans- acknowledged. Support in the form of Research

Seminars in Liver Disease Vol. 33 No. 1/2013


HEV Virology Holla et al 11

Fellowships (to IA and RPH) from the Council for Scientic 21 Reyes GR, Purdy MA, Kim JP, et al. Isolation of a cDNA from the
and Industrial Research, India, is also acknowledged. virus responsible for enterically transmitted non-A, non-B hepa-
titis. Science 1990;247(4948):13351339
22 Huang CC, Nguyen D, Fernandez J, et al. Molecular cloning and
sequencing of the Mexico isolate of hepatitis E virus (HEV).
References Virology 1992;191(2):550558
23 Panda SK, Ansari IH, Durgapal H, Agrawal S, Jameel S. The in vitro-
1 Emerson SU, Purcell RH. Hepatitis E virus. In: Knipe DM, Howley
synthesized RNA from a cDNA clone of hepatitis E virus is
PM, Grifn DE, Lamb RA, Martin MA, Roizman B, Straus SE, eds.
infectious. J Virol 2000;74(5):24302437
Fields Virology. 5th ed. Philadelphia: Lippincott Williams &
24 Emerson SU, Zhang M, Meng XJ, et al. Recombinant hepatitis E
Wilkins; 200630473058
virus genomes infectious for primates: importance of capping
2 Hepatitis E. Media Centre, World Health Organization. 2012.
and discovery of a cis-reactive element. Proc Natl Acad Sci U S A
Available at: http://www.who.int/mediacentre/factsheets/
2001;98(26):1527015275
fs280/en/. Accessed October 15, 2012
25 Graff J, Torian U, Nguyen H, Emerson SU. A bicistronic subgenomic
3 Khuroo MS. Discovery of hepatitis E: the epidemic non-A, non-B
mRNA encodes both the ORF2 and ORF3 proteins of hepatitis E
hepatitis 30 years down the memory lane. Virus Res 2011;161
virus. J Virol 2006;80(12):59195926
(1):314
26 Huang YW, Opriessnig T, Halbur PG, Meng XJ. Initiation at the
4 Meng XJ, Anderson D, Arankalle VA, et al. Hepeviridae. In: King
third in-frame AUG codon of open reading frame 3 of the hepatitis
AMQ, Adams MJ, Carstens EB, Lefkowitz EJ, eds. Virus Taxonomy,
E virus is essential for viral infectivity in vivo. J Virol 2007;81
Ninth Report of the International Committee on Taxonomy of
(6):30183026
Viruses. Oxford: Elsevier/Academic Press; 2011:10211028
27 Ichiyama K, Yamada K, Tanaka T, et al. Determination of the 5-
5 Okamoto H. Genetic variability and evolution of hepatitis E virus.
terminal sequence of subgenomic RNA of hepatitis E virus strains

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


Virus Res 2007;127(2):216228
in cultured cells. Arch Virol 2009;154(12):19451951
6 Uchida T, Suzuki K, Iida F, et al. Animal model, virology and gene
28 Pelosi E, Clarke I. Hepatitis E: a complex and global disease. Emerg
cloning of hepatitis E. Gastroenterol Jpn 1991;26(Suppl 3):148
Health Threats J 2008;1:e8
151
29 Haqshenas G, Shivaprasad HL, Woolcock PR, Read DH, Meng XJ.
7 Ticehurst J, Rhodes LL Jr, Krawczynski K, et al. Infection of owl
Genetic identication and characterization of a novel virus
monkeys (Aotus trivirgatus) and cynomolgus monkeys (Macaca
related to human hepatitis E virus from chickens with hepati-
fascicularis) with hepatitis E virus from Mexico. J Infect Dis
tis-splenomegaly syndrome in the United States. J Gen Virol
1992;165(5):835845
2001;82(Pt 10):24492462
8 McCaustland KA, Krawczynski K, Ebert JW, et al. Hepatitis E virus
30 Huang FF, Sun ZF, Emerson SU, et al. Determination and analysis
infection in chimpanzees: a retrospective analysis. Arch Virol
of the complete genomic sequence of avian hepatitis E virus
2000;145(9):19091918
(avian HEV) and attempts to infect rhesus monkeys with avian
9 Meng XJ, Halbur PG, Haynes JS, et al. Experimental infection of
HEV. J Gen Virol 2004;85(Pt 6):16091618
pigs with the newly identied swine hepatitis E virus (swine
31 Johne R, Heckel G, Plenge-Bnig A, et al. Novel hepatitis E virus
HEV), but not with human strains of HEV. Arch Virol 1998;143
genotype in Norway rats, Germany. Emerg Infect Dis 2010;16
(7):14051415
(9):14521455
10 Viswanathan R. Infectious hepatitis in Delhi (195556) A critical
32 Raj VS, Smits SL, Pas SD, et al. Novel hepatitis E virus in ferrets, the
study: epidemiology. Indian J Med Res 1957;45(Suppl 1):129
Netherlands. Emerg Infect Dis 2012;18(8):13691370
11 Khuroo MS. Discovery of hepatitis E: the epidemic non-A, non-B
33 Zhao C, Ma Z, Harrison TJ, et al. A novel genotype of hepatitis E
hepatitis 30 years down the memory lane. Virus Res 2011;161
virus prevalent among farmed rabbits in China. J Med Virol
(1):314
2009;81(8):13711379
12 Balayan MS, Andjaparidze AG, Savinskaya SS, et al. Evidence for a
34 Jirintai S, Jinshan, Tanggis, et al. Molecular analysis of hepatitis E
virus in non-A, non-B hepatitis transmitted via the fecal-oral
virus from farm rabbits in Inner Mongolia, China and its success-
route. Intervirology 1983;20(1):2331
ful propagation in A549 and PLC/PRF/5 cells. Virus Res 2012;170
13 Velzquez O, Stetler HC, Avila C, et al. Epidemic transmission of
(12):126137
enterically transmitted non-A, non-B hepatitis in Mexico, 1986-
35 Batts W, Yun S, Hedrick R, Winton J. A novel member of the family
1987. JAMA 1990;263(24):32813285
Hepeviridae from cutthroat trout (Oncorhynchus clarkii). Virus
14 Zhuang H, Cao XY, Liu CB, Wang GM. Epidemiology of hepatitis E
Res 2011;158(12):116123
in China. Gastroenterol Jpn 1991;26(Suppl 3):135138
36 Wang H, Zhang W, Ni B, et al. Recombination analysis reveals a
15 Naik SR, Aggarwal R, Salunke PN, Mehrotra NN. A large water-
double recombination event in hepatitis E virus. Virol J
borne viral hepatitis E epidemic in Kanpur, India. Bull World
2010;7:129
Health Organ 1992;70(5):597604
37 Kamar N, Izopet J, Cintas P, et al. Hepatitis E virus-induced
16 Teshale EH, Howard CM, Grytdal SP, et al. Hepatitis E epidemic,
neurological symptoms in a kidney-transplant patient with
Uganda. Emerg Infect Dis 2010;16(1):126129
chronic hepatitis. Am J Transplant 2010;10(5):13211324
17 Nicand E, Armstrong GL, Enouf V, et al. Genetic heterogeneity of
38 Lhomme S, Abravanel F, Dubois M, et al. Hepatitis E virus
hepatitis E virus in Darfur, Sudan, and neighboring Chad. J Med
quasispecies and the outcome of acute hepatitis E in solid-organ
Virol 2005;77(4):519521
transplant patients. J Virol 2012;86(18):1000610014
18 Tam AW, Smith MM, Guerra ME, et al. Hepatitis E virus (HEV):
39 Shukla P, Nguyen HT, Torian U, et al. Cross-species infections of
molecular cloning and sequencing of the full-length viral ge-
cultured cells by hepatitis E virus and discovery of an infectious
nome. Virology 1991;185(1):120131
virus-host recombinant. Proc Natl Acad Sci U S A 2011;108
19 Reyes GR, Huang CC, Tam AW, Purdy MA. Molecular organization
(6):24382443
and replication of hepatitis E virus (HEV). Arch Virol Suppl
40 Shukla P, Nguyen HT, Faulk K, et al. Adaptation of a genotype 3
1993;7:1525
hepatitis E virus to efcient growth in cell culture depends on an
20 Ahmad I, Holla RP, Jameel S. Molecular virology of hepatitis E
inserted human gene segment acquired by recombination. J Virol
virus. Virus Res 2011;161(1):4758
2012;86(10):56975707

Seminars in Liver Disease Vol. 33 No. 1/2013


12 HEV Virology Holla et al

41 Khudyakov YuE, Favorov MO, Jue DL, Hine TK, Fields HA. Immu- 62 Neuvonen M, Ahola T. Differential activities of cellular and viral
nodominant antigenic regions in a structural protein of the macro domain proteins in binding of ADP-ribose metabolites. J
hepatitis E virus. Virology 1994;198(1):390393 Mol Biol 2009;385(1):212225
42 Panda SK, Nanda SK, Zafrullah M, Ansari IH, Ozdener MH, Jameel 63 Purdy MA, Lara J, Khudyakov YE. The hepatitis E virus polyproline
S. An Indian strain of hepatitis E virus (HEV): cloning, sequence, region is involved in viral adaptation. PLoS ONE 2012;7(4):
and expression of structural region and antibody responses in e35974
sera from individuals from an area of high-level HEV endemicity. J 64 Purdy MA. Evolution of the hepatitis E virus polyproline region:
Clin Microbiol 1995;33(10):26532659 order from disorder. J Virol 2012;86(18):1018610193
43 Koonin EV, Gorbalenya AE, Purdy MA, Rozanov MN, Reyes GR, 65 Jameel S, Zafrullah M, Ozdener MH, Panda SK. Expression in
Bradley DW. Computer-assisted assignment of functional do- animal cells and characterization of the hepatitis E virus struc-
mains in the nonstructural polyprotein of hepatitis E virus: tural proteins. J Virol 1996;70(1):207216
delineation of an additional group of positive-strand RNA plant 66 Zafrullah M, Ozdener MH, Kumar R, Panda SK, Jameel S. Muta-
and animal viruses. Proc Natl Acad Sci U S A 1992;89(17): tional analysis of glycosylation, membrane translocation, and cell
82598263 surface expression of the hepatitis E virus ORF2 protein. J Virol
44 Ropp SL, Tam AW, Beames B, Purdy M, Frey TK. Expression of the 1999;73(5):40744082
hepatitis E virus ORF1. Arch Virol 2000;145(7):13211337 67 Graff J, Zhou YH, Torian U, et al. Mutations within potential
45 Sehgal D, Thomas S, Chakraborty M, Jameel S. Expression and glycosylation sites in the capsid protein of hepatitis E virus
processing of the hepatitis E virus ORF1 nonstructural polypro- prevent the formation of infectious virus particles. J Virol
tein. Virol J 2006;3:38 2008;82(3):11851194
46 Ansari IH, Nanda SK, Durgapal H, et al. Cloning, sequencing, and 68 Yamada K, Takahashi M, Hoshino Y, et al. Construction of an
expression of the hepatitis E virus (HEV) nonstructural open infectious cDNA clone of hepatitis E virus strain JE03-1760F that
reading frame 1 (ORF1). J Med Virol 2000;60(3):275283 can propagate efciently in cultured cells. J Gen Virol 2009;90(Pt

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


47 Ahola T, Kriinen L. Reaction in alphavirus mRNA capping: 2):457462
formation of a covalent complex of nonstructural protein nsP1 69 Jimnez de Oya N, Escribano-Romero E, Blzquez AB, et al.
with 7-methyl-GMP. Proc Natl Acad Sci U S A 1995;92(2): Characterization of hepatitis E virus recombinant ORF2 proteins
507511 expressed by vaccinia viruses. J Virol 2012;86(15):78807886
48 Magden J, Takeda N, Li T, et al. Virus-specic mRNA capping 70 Li TC, Yamakawa Y, Suzuki K, et al. Expression and self-assembly
enzyme encoded by hepatitis E virus. J Virol 2001;75(14): of empty virus-like particles of hepatitis E virus. J Virol 1997;71
62496255 (10):72077213
49 Karpe YA, Lole KS. RNA 5-triphosphatase activity of the hepatitis 71 Robinson RA, Burgess WH, Emerson SU, et al. Structural charac-
E virus helicase domain. J Virol 2010;84(18):96379641 terization of recombinant hepatitis E virus ORF2 proteins in
50 Zhang M, Purcell RH, Emerson SU. Identication of the 5 terminal baculovirus-infected insect cells. Protein Expr Purif 1998;12
sequence of the SAR-55 and MEX-14 strains of hepatitis E virus (1):7584
and conrmation that the genome is capped. J Med Virol 2001;65 72 Xing L, Kato K, Li T, et al. Recombinant hepatitis E capsid protein
(2):293295 self-assembles into a dual-domain T 1 particle presenting
51 Pichlmair A, Schulz O, Tan CP, et al. RIG-I-mediated antiviral native virus epitopes. Virology 1999;265(1):3545
responses to single-stranded RNA bearing 5-phosphates. Science 73 Li SW, Zhang J, He ZQ, et al. Mutational analysis of essential
2006;314(5801):9971001 interactions involved in the assembly of hepatitis E virus capsid. J
52 Suppiah S, Zhou Y, Frey TK. Lack of processing of the expressed Biol Chem 2005;280(5):34003406
ORF1 gene product of hepatitis E virus. Virol J 2011;8:245 74 Yamashita T, Mori Y, Miyazaki N, et al. Biological and immuno-
53 Karpe YA, Lole KS. Deubiquitination activity associated with logical characteristics of hepatitis E virus-like particles based on
hepatitis E virus putative papain-like cysteine protease. J Gen the crystal structure. Proc Natl Acad Sci U S A 2009;106
Virol 2011;92(Pt 9):20882092 (31):1298612991
54 Karpe YA, Meng XJ. Hepatitis E virus replication requires an active 75 Guu TS, Liu Z, Ye Q, et al. Structure of the hepatitis E virus-like
ubiquitin-proteasome system. J Virol 2012;86(10):59485952 particle suggests mechanisms for virus assembly and receptor
55 Karpe YA, Lole KS. NTPase and 5 to 3 RNA duplex-unwinding binding. Proc Natl Acad Sci U S A 2009;106(31):1299212997
activities of the hepatitis E virus helicase domain. J Virol 2010;84 76 Mori Y, Matsuura Y. Structure of hepatitis E viral particle. Virus
(7):35953602 Res 2011;161(1):5964
56 Agrawal S, Gupta D, Panda SK. The 3 end of hepatitis E virus (HEV) 77 Zhang JZ, Ng MH, Xia NS, et al. Conformational antigenic deter-
genome binds specically to the viral RNA-dependent RNA minants generated by interactions between a bacterially ex-
polymerase (RdRp). Virology 2001;282(1):87101 pressed recombinant peptide of the hepatitis E virus structural
57 Rehman S, Kapur N, Durgapal H, Panda SK. Subcellular localiza- protein. J Med Virol 2001;64(2):125132
tion of hepatitis E virus (HEV) replicase. Virology 2008;370 78 Li SW, Zhang J, Li YM, et al. A bacterially expressed particulate
(1):7792 hepatitis E vaccine: antigenicity, immunogenicity and protectiv-
58 Aguiar RC, Takeyama K, He C, Kreinbrink K, Shipp MA. B-aggres- ity on primates. Vaccine 2005;23(22):28932901
sive lymphoma family proteins have unique domains that mod- 79 Li S, Tang X, Seetharaman J, et al. Dimerization of hepatitis E virus
ulate transcription and exhibit poly(ADP-ribose) polymerase capsid protein E2s domain is essential for virus-host interaction.
activity. J Biol Chem 2005;280(40):3375633765 PLoS Pathog 2009;5(8):e1000537
59 Salonen A, Ahola T, Kriinen L. Viral RNA replication in 80 Schoeld DJ, Glamann J, Emerson SU, Purcell RH. Identication by
association with cellular membranes. Curr Top Microbiol Immu- phage display and characterization of two neutralizing chimpan-
nol 2005;285:139173 zee monoclonal antibodies to the hepatitis E virus capsid protein.
60 Snijder EJ, Bredenbeek PJ, Dobbe JC, et al. Unique and conserved J Virol 2000;74(12):55485555
features of genome and proteome of SARS-coronavirus, an early 81 Meng J, Dai X, Chang JC, et al. Identication and characterization
split-off from the coronavirus group 2 lineage. J Mol Biol of the neutralization epitope(s) of the hepatitis E virus. Virology
2003;331(5):9911004 2001;288(2):203211
61 Egloff MP, Malet H, Putics A, et al. Structural and functional basis 82 Zhou YH, Purcell RH, Emerson SU. An ELISA for putative neutral-
for ADP-ribose and poly(ADP-ribose) binding by viral macro izing antibodies to hepatitis E virus detects antibodies to geno-
domains. J Virol 2006;80(17):84938502 types 1, 2, 3, and 4. Vaccine 2004;22(20):25782585

Seminars in Liver Disease Vol. 33 No. 1/2013


HEV Virology Holla et al 13

83 Shrestha MP, Scott RM, Joshi DM, et al. Safety and efcacy of a 103 Chandra V, Kalia M, Hajela K, Jameel S. The ORF3 protein of
recombinant hepatitis E vaccine. N Engl J Med 2007;356(9):895903 hepatitis E virus delays degradation of activated growth factor
84 Zhu FC, Zhang J, Zhang XF, et al. Efcacy and safety of a recombi- receptors by interacting with CIN85 and blocking formation of
nant hepatitis E vaccine in healthy adults: a large-scale, random- the Cbl-CIN85 complex. J Virol 2010;84(8):38573867
ised, double-blind placebo-controlled, phase 3 trial. Lancet 104 Taneja S, Sen S, Gupta VK, Aggarwal R, Jameel S. Plasma and
2010;376(9744):895902 urine biomarkers in acute viral hepatitis E. Proteome Sci 2009;
85 Xing L, Wang JC, Li TC, et al. Spatial conguration of hepatitis E 7:39
virus antigenic domain. J Virol 2011;85(2):11171124 105 Tyagi S, Surjit M, Roy AK, Jameel S, Lal SK. The ORF3 protein of
86 Li TC, Suzaki Y, Ami Y, Dhole TN, Miyamura T, Takeda N. Protec- hepatitis E virus interacts with liver-specic alpha1-microglobu-
tion of cynomolgus monkeys against HEV infection by oral lin and its precursor alpha1-microglobulin/bikunin precursor
administration of recombinant hepatitis E virus-like particles. (AMBP) and expedites their export from the hepatocyte. J Biol
Vaccine 2004;22(3-4):370377 Chem 2004;279(28):2930829319
87 Sanford BJ, Opriessnig T, Kenney SP, Dryman BA, Crdoba L, Meng 106 Tyagi S, Surjit M, Lal SK. The 41-amino-acid C-terminal region of
XJ. Assessment of the cross-protective capability of recombinant the hepatitis E virus ORF3 protein interacts with bikunin, a
capsid proteins derived from pig, rat, and avian hepatitis E viruses kunitz-type serine protease inhibitor. J Virol 2005;79
(HEV) against challenge with a genotype 3 HEV in pigs. Vaccine (18):1208112087
2012;30(44):62496255 107 Ratra R, Kar-Roy A, Lal SK. The ORF3 protein of hepatitis E virus
88 He S, Miao J, Zheng Z, et al. Putative receptor-binding sites of interacts with hemopexin by means of its 26 amino acid N-
hepatitis E virus. J Gen Virol 2008;89(Pt 1):245249 terminal hydrophobic domain II. Biochemistry 2008;47
89 Zheng ZZ, Miao J, Zhao M, et al. Role of heat-shock protein 90 in (7):19571969
hepatitis E virus capsid trafcking. J Gen Virol 2010;91(Pt 108 Ratra R, Kar-Roy A, Lal SK. ORF3 protein of hepatitis E virus
7):17281736 interacts with the Bbeta chain of brinogen resulting in de-

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.


90 Kalia M, Chandra V, Rahman SA, Sehgal D, Jameel S. Heparan creased brinogen secretion from HuH-7 cells. J Gen Virol
sulfate proteoglycans are required for cellular binding of the 2009;90(Pt 6):13591370
hepatitis E virus ORF2 capsid protein and for viral infection. J 109 Surjit M, Oberoi R, Kumar R, Lal SK. Enhanced alpha1 micro-
Virol 2009;83(24):1271412724 globulin secretion from hepatitis E virus ORF3-expressing human
91 Kalia M, Jameel S. Virus entry paradigms. Amino Acids 2011;41 hepatoma cells is mediated by the tumor susceptibility gene 101.
(5):11471157 J Biol Chem 2006;281(12):81358142
92 Surjit M, Jameel S, Lal SK. Cytoplasmic localization of the ORF2 110 Moin SM, Chandra V, Arya R, Jameel S. The hepatitis E virus ORF3
protein of hepatitis E virus is dependent on its ability to undergo protein stabilizes HIF-1alpha and enhances HIF-1-mediated tran-
retrotranslocation from the endoplasmic reticulum. J Virol scriptional activity through p300/CBP. Cell Microbiol 2009;11
2007;81(7):33393345 (9):14091421
93 John L, Thomas S, Herchenrder O, Ptzer BM, Schaefer S. 111 Chandra V, Holla P, Ghosh D, Chakrabarti D, Padigaru M, Jameel S.
Hepatitis E virus ORF2 protein activates the pro-apoptotic gene The hepatitis E virus ORF3 protein regulates the expression of
CHOP and anti-apoptotic heat shock proteins. PLoS ONE 2011;6 liver-specic genes by modulating localization of hepatocyte
(9):e25378 nuclear factor 4. PLoS ONE 2011;6(7):e22412
94 Surjit M, Varshney B, Lal SK. The ORF2 glycoprotein of hepatitis E 112 Takahashi M, Yamada K, Hoshino Y, et al. Monoclonal antibodies
virus inhibits cellular NF-B activity by blocking ubiquitination raised against the ORF3 protein of hepatitis E virus (HEV) can
mediated proteasomal degradation of IB in human hepatoma capture HEV particles in culture supernatant and serum but not
cells. BMC Biochem 2012;13(1):7 those in feces. Arch Virol 2008;153(9):17031713
95 Emerson SU, Nguyen H, Torian U, Purcell RH. ORF3 protein of 113 Emerson SU, Nguyen HT, Torian U, Burke D, Engle R, Purcell RH.
hepatitis E virus is not required for replication, virion assembly, Release of genotype 1 hepatitis E virus from cultured hepatoma
or infection of hepatoma cells in vitro. J Virol 2006;80 and polarized intestinal cells depends on open reading frame 3
(21):1045710464 protein and requires an intact PXXP motif. J Virol 2010;84
96 Graff J, Nguyen H, Yu C, et al. The open reading frame 3 gene of (18):90599069
hepatitis E virus contains a cis-reactive element and encodes a 114 Nagashima S, Takahashi M, Jirintai, et al. A PSAP motif in the ORF3
protein required for infection of macaques. J Virol 2005;79 protein of hepatitis E virus is necessary for virion release from
(11):66806689 infected cells. J Gen Virol 2011;92(Pt 2):269278
97 Zafrullah M, Ozdener MH, Panda SK, Jameel S. The ORF3 protein of 115 Nagashima S, Takahashi M, Jirintai S, et al. Tumour susceptibility
hepatitis E virus is a phosphoprotein that associates with the gene 101 and the vacuolar protein sorting pathway are required
cytoskeleton. J Virol 1997;71(12):90459053 for the release of hepatitis E virions. J Gen Virol 2011;92(Pt
98 Chandra V, Kar-Roy A, Kumari S, Mayor S, Jameel S. The hepatitis E 12):28382848
virus ORF3 protein modulates epidermal growth factor receptor 116 Kenney SP, Pudupakam RS, Huang YW, Pierson FW, LeRoith T,
trafcking, STAT3 translocation, and the acute-phase response. J Meng XJ. The PSAP motif within the ORF3 protein of an avian
Virol 2008;82(14):71007110 strain of the hepatitis E virus is not critical for viral infectivity in
99 Kannan H, Fan S, Patel D, Bossis I, Zhang YJ. The hepatitis E virus vivo but plays a role in virus release. J Virol 2012;86(10):5637
open reading frame 3 product interacts with microtubules and 5646
interferes with their dynamics. J Virol 2009;83(13):63756382 117 Chen BJ, Lamb RA. Mechanisms for enveloped virus budding: can
100 Kar-Roy A, Korkaya H, Oberoi R, Lal SK, Jameel S. The hepatitis E some viruses do without an ESCRT? Virology 2008;372(2):221
virus open reading frame 3 protein activates ERK through binding 232
and inhibition of the MAPK phosphatase. J Biol Chem 2004;279 118 Reyes GR, Huang CC, Tam AW, Purdy MA. Molecular organization
(27):2834528357 and replication of hepatitis E virus (HEV). Arch Virol Suppl
101 Korkaya H, Jameel S, Gupta D, et al. The ORF3 protein of hepatitis E 1993;7:1525
virus binds to Src homology 3 domains and activates MAPK. J Biol 119 Kapur N, Thakral D, Durgapal H, Panda SK. Hepatitis E virus enters
Chem 2001;276(45):4238942400 liver cells through receptor-dependent clathrin-mediated endo-
102 Moin SM, Panteva M, Jameel S. The hepatitis E virus Orf3 protein cytosis. J Viral Hepat 2012;19(6):436448
protects cells from mitochondrial depolarization and death. J Biol 120 Nanda SK, Panda SK, Durgapal H, Jameel S. Detection of the
Chem 2007;282(29):2112421133 negative strand of hepatitis E virus RNA in the livers of

Seminars in Liver Disease Vol. 33 No. 1/2013


14 HEV Virology Holla et al

experimentally infected rhesus monkeys: evidence for viral 125 Kamar N, Rostaing L, Abravanel F, et al. Ribavirin therapy inhibits
replication. J Med Virol 1994;42(3):237240 viral replication on patients with chronic hepatitis e virus infec-
121 Williams TP, Kasorndorkbua C, Halbur PG, et al. Evidence of tion. Gastroenterology 2010;139(5):16121618
extrahepatic sites of replication of the hepatitis E virus in a swine 126 de Niet A, Zaaijer HL, ten Berge I, Weegink CJ, Reesink HW, Beuers
model. J Clin Microbiol 2001;39(9):30403046 U. Chronic hepatitis E after solid organ transplantation. Neth J
122 Ippagunta SK, Naik S, Jameel S, Ramana KN, Aggarwal R. Viral RNA Med 2012;70(6):261266
but no evidence of replication can be detected in the peripheral 127 Pischke S, Stiefel P, Franz B, et al. Chronic hepatitis e in
blood mononuclear cells of hepatitis E virus-infected patients. J heart transplant recipients. Am J Transplant 2012;12(11):
Viral Hepat 2011;18(9):668672 31283133
123 Sriram B, Thakral D, Panda SK. Targeted cleavage of hepatitis E 128 Gerolami R, Borentain P, Raissouni F, Motte A, Solas C, Colson P.
virus 3 end RNA mediated by hammerhead ribozymes inhibits Treatment of severe acute hepatitis E by ribavirin. J Clin Virol
viral RNA replication. Virology 2003;312(2):350358 2011;52(1):6062
124 Huang F, Zhou J, Yang Z, et al. RNA interference inhibits hepatitis E 129 Goyal R, Kumar A, Panda SK, Paul SB, Acharya SK. Ribavirin
virus mRNA accumulation and protein synthesis in vitro. Vet therapy for hepatitis E virus-induced acute on chronic liver
Microbiol 2010;142(3-4):261267 failure: a preliminary report. Antivir Ther 2012;17(6):10911096

Downloaded by: Scott Memorial Library, AISR. Copyrighted material.

Seminars in Liver Disease Vol. 33 No. 1/2013

Das könnte Ihnen auch gefallen