Next Article in Journal
Development and Validation of a New DIVA Real-Time PCR Allowing to Differentiate Wild-Type Lumpy Skin Disease Virus Strains, Including the Asian Recombinant Strains, from Neethling-Based Vaccine Strains
Previous Article in Journal
Genome Characterization and Infectivity Potential of Vibriophage-ϕLV6 with Lytic Activity against Luminescent Vibrios of Penaeus vannamei Shrimp Aquaculture
Previous Article in Special Issue
Co-Infection of the Epstein–Barr Virus and the Kaposi Sarcoma-Associated Herpesvirus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Expanding the Hepatitis E Virus Toolbox: Selectable Replicons and Recombinant Reporter Genomes

by
Noémie Oechslin
,
Maliki Ankavay
,
Darius Moradpour
and
Jérôme Gouttenoire
*
Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2023, 15(4), 869; https://doi.org/10.3390/v15040869
Submission received: 6 March 2023 / Revised: 27 March 2023 / Accepted: 28 March 2023 / Published: 28 March 2023
(This article belongs to the Special Issue State-of-the-Art Virology Research in Switzerland)

Abstract

:
Hepatitis E virus (HEV) has received relatively little attention for decades although it is now considered as one of the most frequent causes of acute hepatitis worldwide. Our knowledge of this enterically-transmitted, positive-strand RNA virus and its life cycle remains scarce but research on HEV has gained momentum more recently. Indeed, advances in the molecular virology of hepatitis E, including the establishment of subgenomic replicons and infectious molecular clones, now allow study of the entire viral life cycle and to explore host factors required for productive infection. Here, we provide an overview on currently available systems, with an emphasis on selectable replicons and recombinant reporter genomes. Furthermore, we discuss the challenges in developing new systems which should enable to further investigate this widely distributed and important pathogen.

1. Introduction

The hepatitis E virus (HEV) was discovered in 1983 as a causative agent of acute hepatitis among Soviet soldiers in Afghanistan [1] and has been molecularly cloned in 1990 [2]. It has been classified as the sole member of the Hepeviridae family. With advanced sequencing technologies and exploration of samples from diverse origins, this family expanded rapidly and is now divided into two subfamilies, the Orthohepevirinae and the Parahepevirinae, of which the latter comprises only the Piscihepevirus genus, corresponding to viruses infecting fish [3] (taxonomy available at https://ictv.global/report/chapter/hepeviridae/hepeviridae (accessed on 1 March 2023)). The subfamily Orthohepevirinae comprises the genera Avihepevirus (bird viruses), Chirohepevirus (bat viruses), Rocahepevirus (rodent viruses), and Paslahepevirus (mammalian viruses). The species Paslahepevirus balayani is composed of eight HEV genotypes of which HEV-1 to HEV-4 represent the most important human pathogens (Figure 1A). However, recent observations indicate that infection of humans can also occur by other viruses including camel HEV-7 [4] or the more distantly related rat HEV ([5,6] and refs. therein).
HEV infection of humans causes mostly self-limiting acute hepatitis which can be accompanied by jaundice. HEV-1 and -2, which are transmitted from humans to humans via the fecal–oral route in resource-limited settings with poor sanitation, are highly pathogenic and may lead to severe hepatitis with up to 25% fatality rate in pregnant women [7,8]. Infections with HEV-3 and -4, which represent a zoonosis transmitted primarily through the consumption of undercooked pork or game meat, are often asymptomatic but can cause symptomatic acute hepatitis, especially in middle-aged and elderly men, trigger acute-on-chronic liver failure or be associated with neurologic, renal or other extrahepatic manifestations [9,10]. HEV-3, as well as HEV-3ra, HEV-4, HEV-7, and rat HEV, may persist in immunocompromised patients and cause chronic hepatitis leading to cirrhosis and liver failure [11].
HEV harbors a 7.2-kb positive-strand RNA genome resembling a cellular mRNA with a m7G cap and a poly-A tail (Figure 1B). The genome encodes three main open reading frames (ORF) and, likely, a fourth, in the case of HEV-1 [12]. ORF1, which is first translated upon infection, encodes the so-called replicase, a large, multifunctional protein harboring methyltransferase (MeT), macro, helicase (Hel), and RNA-dependent RNA polymerase (RdRp) domains (Figure 1B). While MeT, Hel, and RdRp have intrinsic roles in viral RNA synthesis, including capping, macro acts as an enzyme known to remove ADP-ribosylation from the posttranslationally modified proteins [13]. A papain-like cysteine protease (PCP) domain has been identified by sequence homology but has not been functionally confirmed yet [14]. A domain with a high-sequence variability known as the hypervariable region (HVR) is located in the middle of ORF1 but has no recognized function. The partially overlapping ORF2 and ORF3, encoded by the last third of the genome, are translated from a subgenomic RNA produced by the viral RdRp (Figure 1B). ORF2 encodes the viral capsid protein which can be produced as infectious, glycosylated, and cleaved forms, the latter supposedly serving as immunologic decoys [15,16]. ORF3 encodes a small, palmitoylated protein which is essential for the secretion of infectious virions as quasi-enveloped particles wrapped in exosomal membranes [17,18].
Not the least due to the limited availability of model systems, knowledge of the HEV life cycle remains relatively scarce. The first functional molecular clones of HEV-1, i.e., the pSGI-HEV(I) [19] and Sar55 clones [20], were described in the early 2000s. Molecular clones of HEV-3 have been described more recently and found to replicate more efficiently in cell culture. They include the p6 clone (Kernow-C1 strain, HEV-3a) [21], the 83-2 clone (HEV83-2-27 strain, HEV-3k) [22], and the pJE03-1760F clone (HEV-3b) [23] (reviewed in [24]) (Figure 1A). An increasing number of molecular clones, including for other genotypes, such as HEV-4 [25], HEV-5 [26], HEV-7 [27], and HEV-8 [28], have been described recently but have been less studied yet.
Studies of viral life cycles have been facilitated by the use of recombinant constructs allowing the expression of a reporter gene. These tools have been key in a number of discoveries, including the receptor used for virus entry or the identification of potent antivirals [29,30]. This strategy has been successfully applied to positive-strand RNA viruses from different families, including, among others, corona- [31,32], alpha- (e.g., Venezuelian equine encephalitis virus [29]), flavi- (reviewed in [33]) and hepaciviruses (hepatitis C virus [34], reviewed in [35]).
Here, we provide an overview on currently available HEV clones, including subgenomic replicons and full-length genomes, which have been engineered to allow selection or reporter expression to study the viral life cycle. Applications and expected values of their use shall be discussed together with their limitations. Furthermore, perspectives and challenges for the development of new HEV constructs shall be addressed.

2. Subgenomic Replicons as Tools to Study HEV RNA Replication

Subgenomic replicons, i.e., constructs comprising the genome elements and allowing expression of the viral machinery required for RNA replication without infectious particle production, have been established for numerous positive-, but also negative-strand, RNA viruses (reviewed in [36]). They allow the study of genuine viral RNA replication and enable manipulation under biosafety level 2 conditions. Thus, replicons have been widely used to study pathogenic RNA viruses and were even instrumental for the development of antiviral therapy against HCV [37].
The first subgenomic HEV replicon was described in 2004 by Emerson et al. [38]. A green fluorescent protein (GFP) sequence with two stop codons was inserted after the methionine start codon of ORF2 and replaced part of the ORF2 and most of the ORF3 sequences, while ORF1 was left unmodified. In this setting, the replicon allows for expression of the entire replicase, the first 14 amino acids (aa) of ORF3 and the GFP reporter but no ORF2 protein is produced. While ORF2 expression is not necessary for viral RNA replication, structural RNA elements, namely, internal stem loops 1 and 2 in the middle of its coding region, were shown later to be essential for efficient HEV RNA replication [39]. Therefore, HEV replicons should preserve these structures as well as some others described at the 3′-end of ORF2 [40]. Thanks to this first HEV replicon, it was possible to monitor HEV replication in cell culture by microscopy or fluorescence-activated cell sorting (FACS). The replicon was further improved with the more stable enhanced GFP [41] but, although it represents a powerful tool to identify HEV-replicating cells after replicon transfection, it does not allow for quantitative assessment of viral replication kinetics.
Quantitative evaluation of viral RNA replication can be achieved by the use of a luciferase reporter (Table 1). Replicons harboring the non-secreted Renilla [42,43] or firefly luciferase [44] have been described. However, the most widely used replicon harbors a Gaussia luciferase (Gluc) sequence inserted downstream of the ORF2 start codon, replacing part of its sequence [21]. Gluc possesses a signal sequence resulting in its secretion and accumulation in the cell culture supernatant. It allows the study of sequential time points in the same transfected cell population and, therefore, to follow replication kinetics over several days. The Gluc replicon has served many purposes, including drug screening [45], mutational [46], and domain swapping analyses [47]. Although very convenient, this reporter is limited by the fact that any defect in the secretory pathway may falsely point toward an impaired replication capacity.
Selection of a cell population which autonomously replicates subgenomic HEV RNA is another interest achievable with replicons. Similar to replicons harboring GFP or luciferase reporters, an antibiotic selection cassette, e.g., the neomycin phosphotransferase II gene (Neo), can be inserted at the start codon of ORF2 (Table 1). The selection of cells replicating an HEV-Neo replicon may serve the study of the mechanisms of RNA production [48] or to evaluate antiviral agents [49]. Beyond these applications, selectable replicons allow the performance of reverse genetics studies taking advantage of the intrinsic rapid evolution of RNA viruses and the emergence of adaptive second-site changes. Selectable subgenomic HEV replicons allowed our laboratory to conduct a transposon-mediated random insertion screen to identify viable insertion sites within the ORF1 region [50] and to select for adaptive changes after site-directed mutagenesis of a conserved α-helix in the RdRp, revealing a functional interaction between the thumb and palm subdomains [51]. While selectable replicons can be a useful tool to identify mutations conferring antiviral resistance [52], they have not been used successfully for this purpose in HEV thus far.
Table 1. Reporter and selection genes used in HEV replicon constructs. The reference corresponding to the first description of the replicon construct is indicated. BSR, Blasticidin-S resistance; Fluc, Firefly luciferase; GFP, Green fluorescent protein; Gluc, Gaussia luciferase; Neo, Neomycin phosphotransferase II; Rluc, Renilla luciferase; Zeo, Zeocin resistance cassette; and ZsGreen, Zoanthus green fluorescent protein.
Table 1. Reporter and selection genes used in HEV replicon constructs. The reference corresponding to the first description of the replicon construct is indicated. BSR, Blasticidin-S resistance; Fluc, Firefly luciferase; GFP, Green fluorescent protein; Gluc, Gaussia luciferase; Neo, Neomycin phosphotransferase II; Rluc, Renilla luciferase; Zeo, Zeocin resistance cassette; and ZsGreen, Zoanthus green fluorescent protein.
Reporter/
Selection Gene
Size
(bp)
ApplicationRef.
Gluc555Replication kinetics[21]
GFP720Evaluation replication level/cell sorting[38]
Neo795Selection of a cell population[48]
Rluc932Replication kinetics[42]
GFP-Zeo1077Selection and eval. replication level [53]
BSR-2A-ZsGreen1153Selection and eval. replication level[54]
Gluc-2A-Neo1408Selection and replication kinetics[55]
Fluc1659Replication kinetics[44]
Selection markers have also been used in combination with fluorescent proteins, either by direct fusion of the reporters, i.e., GFP-Zeo [53], or by the insertion of the self-cleaving 2A peptide, i.e., BSR-2A-ZsGreen [54] (Table 1). These constructs allow, first, the selection of a population of cells stably replicating HEV and, then, the analysis of the effect of various drugs by evaluating the fluorescence intensity at the single-cell level [53,54]. Similarly, a combination of Gluc with a selection marker has recently been described, i.e., Gluc-2A-Neo, enabling monitoring of viral replication by a simple luciferase activity measurement in the supernatant of a homogenous cell population harboring replicating HEV [55].Interestingly, the strategy consisting in expressing a reporter in place of ORF2 and ORF3 can be applied to different HEV genotypes. Therefore, Gluc or Neo replicons have been prepared, in addition to HEV-1 and -3, for HEV-4 [25], HEV-5 [26], HEV-7 [27], and HEV-8 [28], but also for the more distant rat HEV [56].
In conclusion, the replicon constructs allow for working with a non-infectious system and offer a versatile platform for the incorporation of a wide range of reporters. While this system has contributed to an improved understanding of HEV RNA replication from the virus and the host cell perspective, one should keep in mind that expression of a reporter gene inserted in place of the ORF2 sequence is linked to the synthesis of the subgenomic HEV RNA. Therefore, any mutation which would affect the generation of this RNA species but not of the full-length RNA may result in misinterpretation as a replication defect. In addition, participation of the ORF2 and ORF3 proteins in RNA replication cannot be evaluated. The recent description of an ORF2 single nucleotide variant which may affect viral RNA replication [57] is supporting this concern. Hence, results obtained in a subgenomic replicon system should be validated in a full-length HEV RNA replication and/or infection system.
Trans-complementation systems have also been developed for HEV. In such systems, subgenomic replicons are transfected into cells expressing in trans the ORF2 and/or ORF3 proteins or co-transfected with full-length viral RNA into naïve cells, in order to produce virus-like particles containing the reporter genome or to restore a defective replicase. These systems allow for uncoupling the RNA replication from assembly and genome packaging, which are both still poorly characterized. The first trans-complementation system was developed by transfection of a Gluc replicon into cells stably expressing ORF2 and/or ORF3 [58]. Viral-like particles were successfully produced and secreted into the cell culture supernatant, allowing study of the role of ORF3 protein in virus secretion [58].
A similar system has been established by the same group to study the mechanisms leading to subgenomic RNA synthesis [59]. Here, cells overexpressing wild-type or polymerase-deficient ORF1 protein were transfected with a Gluc construct harboring a deletion in the ORF1 sequence, allowing the mapping of the intragenomic subgenomic promoter [59]. Moreover, a trans-complementation system was also used to package a GFP replicon into viral particles by co-transfection with a full-length RNA genome. In this setting viral particles were infectious, but not all of them delivered a GFP replicon after infection [60]. While trans-complementation assays leading to encapsidation of a replicon construct offer interesting perspectives, such as lowering the biosafety level required for in vitro infection compared to the use of authentic infectious clones, they are strongly hampered by their relative inefficiency in producing virus-like particles. This illustrates the need to improve trans-complementation assays but also to develop full-length HEV genomes harboring reporters.

3. Infectious HEV Clones Harboring Reporters

Viruses are under constant selection pressure, forming the basis for their evolution and optimization of genome size. Therefore, viral genomes offer little room to accommodate reporter sequences or entire genes. Moreover, in the case of HEV, available model systems produce relatively low infectious titers as compared to some other viruses. Therefore, identifying viable insertion sites to introduce a reporter gene with minimal impact on replication is challenging. Two approaches can be taken, either by unbiased random insertion and selection for viable insertion sites or by targeting regions of the genome that are less well conserved and, potentially, more tolerant to foreign sequence insertion.
Based on previous work on the hepatitis C virus [61], an unbiased approach to identify viable insertion sites in HEV ORF1 was recently taken by our laboratory. Transposon-mediated random insertion of a 15-nt (5-aa) sequence in a selectable subgenomic replicon (83-2 clone) allowed the identification of viable insertion sites in the MeT, the HVR, and between the Hel and RdRp domains [50]. When the 5-aa transposon sequence was replaced by the larger HA epitope tag, only insertions in the HVR retained full capacity to replicate and to produce infectious virus (Figure 2). These HA-tagged genomes allowed for the convenient and highly reproducible detection of the HEV replicase by immunoblot as well as immunofluorescence, demonstrating that the largely predominant form of ORF1 protein corresponds to the unprocessed polyprotein of ~200 kDa and yielding first insights into putative viral replication sites. Of note, genomes harboring a GFP sequence insertion were found to be replication competent but did not yield any visible fluorescent signal, indicating that reporters of up to 25 kDa may be tolerated in the HVR (unpublished data). Furthermore, we have shown that insertion of NanoLuc flanked by minimal linker sequences is viable (Figure 2). While RNA replication capacity appears reduced as compared to the parental HEV 83-2 clone, the inserted enzyme remained fully functional and allowed for the quantitative assessment of viral replication kinetics. Moreover, a full-length genome harboring NanoLuc in the identified site produced infectious virus, however, at a lower titer than the parental clone [50]. Hence, luciferase activity can serve to quantify viral replication after genuine HEV infection, facilitating future studies on viral entry or antiviral drug discovery efforts.
HEV presents relatively high genetic variability, with ≥25% nucleotide sequence diversity between major human-pathogenic genotypes 1–4. Important intra-genotype variability is also observed, especially in HEV-3, with particularly high sequence diversity in the so-called hypervariable region (HVR) of ORF1 (Figure 2). Of note, HEV-3ra, known to infect rabbits as natural host, usually harbors a highly variable 31-aa (93-nucleotide (nt)) insertion between the macro and helicase domains [62,63,64].
Interestingly, HEV-3 genomes isolated from patients with chronic hepatitis E sometimes harbor insertions of different lengths in the HVR, ranging from 60 to 333 nt ([65,66] and reviewed in [67]). The nature of the inserted sequence is also highly variable, including duplications of HEV sequences from the HVR or RdRp regions or fragments of human mRNA [65,66,68,69,70,71]. Inserted sequences are often rich in prolines as well as polar residues, which is close to the aa composition found in the targeted HVR sequence (Figure 2). Moreover, the well characterized insertion found in the Kernow-C1 p6 infectious clone has been shown to provide a replication advantage in cell culture [21]. This 174-nt (58-aa) insertion derived from ribosomal protein S17 (RPS17) RNA was selected in cell culture after infection with a stool sample from a patient with chronic hepatitis E [69,72]. Hence, the identification of sequence insertions in the HVR of natural HEV isolates suggests that epitope tags or reporter sequences inserted in this region may be viable.
Taking advantage of HEV-3 sequence diversity, Metzger et al. employed a targeted approach to establish full-length Kernow-C1 p6 genomes harboring hemagglutinin (HA, 9 aa) or V5 (14 aa) epitope tags in the HVR [73], at a site where a naturally occurring insertion had been described [74] (Figure 2). Of note, as the aa sequence of the V5 tag comprises 3 proline residues and partly resembles the targeted HVR sequence, the authors adopted a mixed strategy of aa substitution and insertion to prepare this construct. The shorter HA tag was less tolerated at the same position, highlighting the importance of the nature of the inserted aa sequence. In addition to preserved RNA replication, full-length genomes harboring a V5 tag in the HVR retained the ability to produce infectious virus. Metzger et al. exploited functionality of the tagged genomes investigate potential processing and the subcellular localization of the ORF1 protein by immunoprecipitation, immunoblotting and immunofluorescence analyses using epitope-specific antibodies [73].
Similarly, Primadharsini et al. have inserted NanoKAZ luciferase, also known as NanoLuc (171 aa, 19 kDa), in the HVR region of the pJE03-1760F clone [75]. Three of five constructs prepared yielded measurable luciferase activity, and only one was found to be genetically stable upon passaging (Figure 2). The selected HEV genome harboring NanoLuc was able to produce infectious virus, including quasi-enveloped particles, and enabled a drug screen yielding four compounds with potential antiviral activity against HEV [75].
Figure 2. Engineered epitope tag and reporter insertions in HEV ORF1 genomic region. The sequence alignment of 155 HEV-3 ORF1 amino acid (aa) sequences, found in the databases, with exclusion of HEV-3ra and sequences harboring insertions in the HVR, served as basis for the aa identity sequence profile. Results of this alignment in the HVR, aa 712–787, is provided as an aa logo generated with the WebLogo application [76]. Some of the reported positions where natural insertions occurred, of which some served as reference for the targeted tagging approaches, are indicated by empty arrowheads with the corresponding citations [68,69,74,77,78]. Positions where epitope tags, HA or V5, as well as NanoLuc reporter have been successfully inserted in HEV-3 molecular clones, i.e., 83-2, p6 or pJE03-1760F, are indicated with their reference [50,73,75].
Figure 2. Engineered epitope tag and reporter insertions in HEV ORF1 genomic region. The sequence alignment of 155 HEV-3 ORF1 amino acid (aa) sequences, found in the databases, with exclusion of HEV-3ra and sequences harboring insertions in the HVR, served as basis for the aa identity sequence profile. Results of this alignment in the HVR, aa 712–787, is provided as an aa logo generated with the WebLogo application [76]. Some of the reported positions where natural insertions occurred, of which some served as reference for the targeted tagging approaches, are indicated by empty arrowheads with the corresponding citations [68,69,74,77,78]. Positions where epitope tags, HA or V5, as well as NanoLuc reporter have been successfully inserted in HEV-3 molecular clones, i.e., 83-2, p6 or pJE03-1760F, are indicated with their reference [50,73,75].
Viruses 15 00869 g002
Current evidence points to the ORF1 region as having the greatest potential to accommodate reporter sequences. By contrast, the last third of the viral genome, encoding mainly ORF2, is likely more restrictive to heterologous sequence insertion given the tight and conserved protein–protein interactions required for capsid assembly. Using transposon-mediated random insertion and selection for infectious virus production, we, nevertheless, recently succeeded in identifying viable insertion sites in ORF2 (manuscript in preparation). In addition, Nishiyama et al. have recently reported a functional HEV genome expressing an ORF2 protein with a C-terminal FLAG tag [79]. Using the pJE03-1760F clone as a backbone, a FLAG tag sequence was fused to ORF2, followed by a stop codon and a duplication of the last 60 nt of the ORF2 sequence preceding the authentic stop codon. This sequence duplication circumvents disruption of a crucial cis-acting replication element present in the 3′ ORF2-coding region [40]. Importantly, quasi-enveloped virus-like particles harboring the FLAG-tagged capsid were secreted from transfected cells, as revealed by detergent treatment and immunoprecipitation analyses [79]. Whether these secreted particles as well as the intracellular virions are infectious remains to be clarified.
While recent studies showed that the HEV ORF1 and ORF2 proteins can tolerate the insertion of epitope tags and reporter genes, tagging of the ORF3 protein is likely more challenging, as almost its entire sequence overlaps with ORF2. More work is needed to further characterize and optimize the engineered tagged genomes. Overall, these hold promises for the study of virus entry, replication, and egress but validation of the versatility of the reporter systems as well as improvements in terms of virus production are a priority.

4. Challenges and Future Developments

The development of HEV in vitro infection and replication systems significantly contributed to a recent enhanced understanding of the viral life cycle, including entry, RNA replication, and virus production. Because current systems remain suboptimal, with modest efficacy and slow infectious virus production as well as rather low infectivity, there is not a single consensual HEV model system employed. Indeed, different cell lines, including some of hepatic origin, i.e., Huh-7 cells or sublines derived thereof, PLC/PRF/5, HepG2, and Hep293TT cells, but also A549 cells derived from lung tissue, are widely used to study HEV infection in vitro. Other systems, such as polarized stem cell-derived hepatocyte-like cells [80] or hepatic organoids [81] which are emerging, offer interesting alternatives to cancer cell lines but remain difficult to establish.
Among the infectious HEV clones, the Kernow-C1 p6 clone is the most widely used in cell culture because it produces the highest viral titers. However, it contains an S17 insertion in the HVR which provides a replication advantage, requiring validation of observations in an independent system. Efforts to establish new molecular clones, ideally with improved replication capacity, but also to identify highly permissive cell lines which may recapitulate all steps of the viral life cycle are crucial to the field.
Recent development in molecular biology, such as the CRISPR-Cas9 technology, now allow for the conducting of large genetic screening using pooled genome-wide single-guide RNA (sgRNA) library. This screening approach has proven over the last years to be extremely powerful for virus–host interaction study with numerous articles in the literature (among many others [29,82,83]), however, none concerned HEV. Beside genetic approaches, proteomics has shown to be very complementary to the study of virus–host interactions (reviewed in [84]). Together with improved sensitivity achieved lately with mass spectrometry (MS) detection methods, development of more robust HEV model systems shall serve the exploration of the host proteome involved during the complete viral life cycle.
Live-cell imaging experiments have not been conducted for HEV but may be an important step toward understanding of the leading mechanisms of infection. Engineering new recombinant viruses expressing fluorescently-labeled proteins may serve to follow the dynamic behavior of the HEV proteins during viral replication and, likely, the entire viral life cycle. Live-cell imaging may also include the detection of RNA molecules using insertion of targetable sequences in the viral genome (reviewed in [85]), as shown previously to study the translation and packaging of HIV-1 genome [86].
Reporter systems have proven to be instrumental in molecular virology to investigate viral life cycles, including the virus–host interactions. HEV model systems have undergone essential development in the last decade with the establishment of several infectious clones as well as selectable and reporter systems, which should translate into an improved understanding of the viral life cycle.

Author Contributions

Writing—original draft preparation, N.O., M.A., D.M. and J.G.; writing—review and editing, N.O., M.A., D.M. and J.G. funding acquisition, D.M. and J.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Swiss National Science Foundation (grants 310030_207477 and CRSK-3_190706) as well as the Novartis Foundation for Medical–Biological Research (grant 18C140).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Balayan, M.S.; Andjaparidze, A.G.; Savinskaya, S.S.; Ketiladze, E.S.; Braginsky, D.M.; Savinov, A.P.; Poleschuk, V.F. Evidence for a virus in non-A, non-B hepatitis transmitted via the fecal-oral route. Intervirology 1983, 20, 23–31. [Google Scholar] [CrossRef] [PubMed]
  2. Reyes, G.R.; Purdy, M.A.; Kim, J.P.; Luk, K.C.; Young, L.M.; Fry, K.E.; Bradley, D.W. Isolation of a cDNA from the virus responsible for enterically transmitted non-A, non-B hepatitis. Science 1990, 247, 1335–1339. [Google Scholar] [CrossRef] [PubMed]
  3. Purdy, M.A.; Drexler, J.F.; Meng, X.J.; Norder, H.; Okamoto, H.; Van der Poel, W.H.M.; Reuter, G.; de Souza, W.M.; Ulrich, R.G.; Smith, D.B. ICTV Virus Taxonomy Profile: Hepeviridae 2022. J. Gen. Virol. 2022, 103, 001778. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, G.H.; Tan, B.H.; Teo, E.C.; Lim, S.G.; Dan, Y.Y.; Wee, A.; Aw, P.P.; Zhu, Y.; Hibberd, M.L.; Tan, C.K.; et al. Chronic infection with camelid hepatitis E virus in a liver transplant recipient who regularly consumes camel meat and milk. Gastroenterology 2016, 150, 355–357.e353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rivero-Juarez, A.; Frias, M.; Perez, A.B.; Pineda, J.A.; Reina, G.; Fuentes-Lopez, A.; Freyre-Carrillo, C.; Ramirez-Arellano, E.; Alados, J.C.; Rivero, A.; et al. Orthohepevirus C infection as an emerging cause of acute hepatitis in Spain: First report in Europe. J. Hepatol. 2022, 77, 326–331. [Google Scholar] [CrossRef] [PubMed]
  6. Sridhar, S.; Yip, C.C.Y.; Lo, K.H.Y.; Wu, S.; Situ, J.; Chew, N.F.S.; Leung, K.H.; Chan, H.S.Y.; Wong, S.C.Y.; Leung, A.W.S.; et al. Hepatitis E virus species C Iinfection in humans, Hong Kong. Clin. Infect. Dis. 2022, 75, 288–296. [Google Scholar] [CrossRef] [PubMed]
  7. Nelson, K.E.; Labrique, A.B.; Kmush, B.L. Epidemiology of genotype 1 and 2 hepatitis E virus infections. Cold. Spring Harb. Perspect. Med. 2019, 9, a031732. [Google Scholar] [CrossRef] [PubMed]
  8. Heemelaar, S.; Hangula, A.L.; Chipeio, M.L.; Josef, M.; Stekelenburg, J.; van den Akker, T.H.; Pischke, S.; Mackenzie, S.B.P. Maternal and fetal outcomes of pregnancies complicated by acute hepatitis E and the impact of HIV status: A cross-sectional study in Namibia. Liver Int. 2022, 42, 50–58. [Google Scholar] [CrossRef]
  9. Debing, Y.; Moradpour, D.; Neyts, J.; Gouttenoire, J. Update on hepatitis E virology: Implications for clinical practice. J. Hepatol. 2016, 65, 200–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kamar, N.; Izopet, J.; Pavio, N.; Aggarwal, R.; Labrique, A.; Wedemeyer, H.; Dalton, H.R. Hepatitis E virus infection. Nat. Rev. Dis. Primers 2017, 3, 17086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. European Association for the Study of the Liver. EASL Clinical Practice Guidelines on hepatitis E virus infection. J. Hepatol. 2018, 68, 1256–1271. [Google Scholar] [CrossRef] [PubMed]
  12. Nair, V.P.; Anang, S.; Subramani, C.; Madhvi, A.; Bakshi, K.; Srivastava, A.; Shalimar; Nayak, B.; Ranjith Kumar, C.T.; Surjit, M. Endoplasmic reticulum stress induced synthesis of a novel viral factor mediates efficient replication of genotype-1 hepatitis E virus. PLoS Pathog. 2016, 12, e1005521. [Google Scholar] [CrossRef] [PubMed]
  13. Li, C.; Debing, Y.; Jankevicius, G.; Neyts, J.; Ahel, I.; Coutard, B.; Canard, B. Viral macro domains reverse protein ADP-ribosylation. J. Virol. 2016, 90, 8478–8486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Koonin, E.V.; Gorbalenya, A.E.; Purdy, M.A.; Rozanov, M.N.; Reyes, G.R.; Bradley, D.W. Computer-assisted assignment of functional domains in the nonstructural polyprotein of hepatitis E virus: Delineation of an additional group of positive-strand RNA plant and animal viruses. Proc. Natl. Acad. Sci. USA 1992, 89, 8259–8263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Montpellier, C.; Wychowski, C.; Sayed, I.M.; Meunier, J.C.; Saliou, J.M.; Ankavay, M.; Bull, A.; Pillez, A.; Abravanel, F.; Helle, F.; et al. Hepatitis E virus lifecycle and identification of 3 forms of the ORF2 capsid protein. Gastroenterology 2018, 154, 211–223.e218. [Google Scholar] [CrossRef] [PubMed]
  16. Yin, X.; Ying, D.; Lhomme, S.; Tang, Z.; Walker, C.M.; Xia, N.; Zheng, Z.; Feng, Z. Origin, antigenicity, and function of a secreted form of ORF2 in hepatitis E virus infection. Proc. Natl. Acad. Sci. USA 2018, 115, 4773–4778. [Google Scholar] [CrossRef] [Green Version]
  17. Nagashima, S.; Takahashi, M.; Kobayashi, T.; Tanggis; Nishizawa, T.; Nishiyama, T.; Primadharsini, P.P.; Okamoto, H. Characterization of the quasi-enveloped hepatitis E virus particles released by the cellular exosomal pathway. J. Virol. 2017, 91, e00822-17. [Google Scholar] [CrossRef] [Green Version]
  18. Gouttenoire, J.; Pollan, A.; Abrami, L.; Oechslin, N.; Mauron, J.; Matter, M.; Oppliger, J.; Szkolnicka, D.; Dao Thi, V.L.; van der Goot, F.G.; et al. Palmitoylation mediates membrane association of hepatitis E virus ORF3 protein and is required for infectious particle secretion. PLoS Pathog. 2018, 14, e1007471. [Google Scholar] [CrossRef] [PubMed]
  19. Panda, S.K.; Ansari, I.H.; Durgapal, H.; Agrawal, S.; Jameel, S. The in vitro-synthesized RNA from a cDNA clone of hepatitis E virus is infectious. J. Virol. 2000, 74, 2430–2437. [Google Scholar] [CrossRef] [Green Version]
  20. Emerson, S.U.; Zhang, M.; Meng, X.J.; Nguyen, H.; St Claire, M.; Govindarajan, S.; Huang, Y.K.; Purcell, R.H. Recombinant hepatitis E virus genomes infectious for primates: Importance of capping and discovery of a cis-reactive element. Proc. Natl. Acad. Sci. USA 2001, 98, 15270–15275. [Google Scholar] [CrossRef] [Green Version]
  21. Shukla, P.; Nguyen, H.T.; Faulk, K.; Mather, K.; Torian, U.; Engle, R.E.; Emerson, S.U. Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J. Virol. 2012, 86, 5697–5707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Shiota, T.; Li, T.C.; Yoshizaki, S.; Kato, T.; Wakita, T.; Ishii, K. The hepatitis E virus capsid C-terminal region is essential for the viral life cycle: Implication for viral genome encapsidation and particle stabilization. J. Virol. 2013, 87, 6031–6036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yamada, K.; Takahashi, M.; Hoshino, Y.; Takahashi, H.; Ichiyama, K.; Tanaka, T.; Okamoto, H. Construction of an infectious cDNA clone of hepatitis E virus strain JE03-1760F that can propagate efficiently in cultured cells. J. Gen. Virol. 2009, 90, 457–462. [Google Scholar] [CrossRef] [PubMed]
  24. Meister, T.L.; Bruening, J.; Todt, D.; Steinmann, E. Cell culture systems for the study of hepatitis E virus. Antiviral Res. 2019, 163, 34–49. [Google Scholar] [CrossRef]
  25. Cordoba, L.; Feagins, A.R.; Opriessnig, T.; Cossaboom, C.M.; Dryman, B.A.; Huang, Y.W.; Meng, X.J. Rescue of a genotype 4 human hepatitis E virus from cloned cDNA and characterization of intergenotypic chimeric viruses in cultured human liver cells and in pigs. J. Gen. Virol. 2012, 93, 2183–2194. [Google Scholar] [CrossRef] [Green Version]
  26. Li, T.C.; Bai, H.; Yoshizaki, S.; Ami, Y.; Suzaki, Y.; Doan, Y.H.; Takahashi, K.; Mishiro, S.; Takeda, N.; Wakita, T. Genotype 5 hepatitis E virus produced by a reverse genetics system has the potential for zoonotic infection. Hepatol. Commun. 2019, 3, 160–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Li, T.C.; Zhou, X.; Yoshizaki, S.; Ami, Y.; Suzaki, Y.; Nakamura, T.; Takeda, N.; Wakita, T. Production of infectious dromedary camel hepatitis E virus by a reverse genetic system: Potential for zoonotic infection. J. Hepatol. 2016, 65, 1104–1111. [Google Scholar] [CrossRef]
  28. Zhang, W.; Ami, Y.; Suzaki, Y.; Doan, Y.H.; Takeda, N.; Muramatsu, M.; Li, T.C. Generation of a bactrian camel hepatitis E virus by a reverse genetics system. J. Gen. Virol. 2021, 102, 001618. [Google Scholar] [CrossRef] [PubMed]
  29. Ma, H.; Kim, A.S.; Kafai, N.M.; Earnest, J.T.; Shah, A.P.; Case, J.B.; Basore, K.; Gilliland, T.C.; Sun, C.; Nelson, C.A.; et al. LDLRAD3 is a receptor for Venezuelan equine encephalitis virus. Nature 2020, 588, 308–314. [Google Scholar] [CrossRef]
  30. Carbajo-Lozoya, J.; Muller, M.A.; Kallies, S.; Thiel, V.; Drosten, C.; von Brunn, A. Replication of human coronaviruses SARS-CoV, HCoV-NL63 and HCoV-229E is inhibited by the drug FK506. Virus Res. 2012, 165, 112–117. [Google Scholar] [CrossRef]
  31. Freeman, M.C.; Graham, R.L.; Lu, X.; Peek, C.T.; Denison, M.R. Coronavirus replicase-reporter fusions provide quantitative analysis of replication and replication complex formation. J. Virol. 2014, 88, 5319–5327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. V’Kovski, P.; Gerber, M.; Kelly, J.; Pfaender, S.; Ebert, N.; Braga Lagache, S.; Simillion, C.; Portmann, J.; Stalder, H.; Gaschen, V.; et al. Determination of host proteins composing the microenvironment of coronavirus replicase complexes by proximity-labeling. eLife 2019, 8, e42037. [Google Scholar] [CrossRef]
  33. Baker, C.; Shi, P.Y. Construction of stable reporter flaviviruses and their applications. Viruses 2020, 12, 1082. [Google Scholar] [CrossRef] [PubMed]
  34. Krieger, N.; Lohmann, V.; Bartenschlager, R. Enhancement of hepatitis C virus RNA replication by cell culture-adaptive mutations. J. Virol. 2001, 75, 4614–4624. [Google Scholar] [CrossRef] [Green Version]
  35. Steinmann, E.; Pietschmann, T. Cell culture systems for hepatitis C virus. Curr. Top. Microbiol. Immunol. 2013, 369, 17–48. [Google Scholar] [CrossRef] [PubMed]
  36. Hannemann, H. Viral replicons as valuable tools for drug discovery. Drug Discov. Today 2020, 25, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  37. Lohmann, V. Hepatitis C virus cell culture models: An encomium on basic research paving the road to therapy development. Med. Microbiol. Immunol. 2019, 208, 3–24. [Google Scholar] [CrossRef]
  38. Emerson, S.U.; Nguyen, H.; Graff, J.; Stephany, D.A.; Brockington, A.; Purcell, R.H. In vitro replication of hepatitis E virus (HEV) genomes and of an HEV replicon expressing green fluorescent protein. J. Virol. 2004, 78, 4838–4846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Emerson, S.U.; Nguyen, H.T.; Torian, U.; Mather, K.; Firth, A.E. An essential RNA element resides in a central region of hepatitis E virus ORF2. J. Gen. Virol. 2013, 94, 1468–1476. [Google Scholar] [CrossRef]
  40. Graff, J.; Nguyen, H.; Kasorndorkbua, C.; Halbur, P.G.; St Claire, M.; Purcell, R.H.; Emerson, S.U. In vitro and in vivo mutational analysis of the 3’-terminal regions of hepatitis E virus genomes and replicons. J. Virol. 2005, 79, 1017–1026. [Google Scholar] [CrossRef] [Green Version]
  41. Pudupakam, R.S.; Huang, Y.W.; Opriessnig, T.; Halbur, P.G.; Pierson, F.W.; Meng, X.J. Deletions of the hypervariable region (HVR) in open reading frame 1 of hepatitis E virus do not abolish virus infectivity: Evidence for attenuation of HVR deletion mutants in vivo. J. Virol. 2009, 83, 384–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Cao, D.; Huang, Y.W.; Meng, X.J. The nucleotides on the stem-loop RNA structure in the junction region of the hepatitis E virus genome are critical for virus replication. J. Virol. 2010, 84, 13040–13044. [Google Scholar] [CrossRef] [Green Version]
  43. Cao, D.; Ni, Y.Y.; Meng, X.J. Substitution of amino acid residue V1213 in the helicase domain of the genotype 3 hepatitis E virus reduces virus replication. Virol. J. 2018, 15, 32. [Google Scholar] [CrossRef] [Green Version]
  44. Netzler, N.E.; Enosi Tuipulotu, D.; Vasudevan, S.G.; Mackenzie, J.M.; White, P.A. Antiviral candidates for treating hepatitis E virus infection. Antimicrob. Agents Chemother. 2019, 63, e00003-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Todt, D.; Francois, C.; Anggakusuma; Behrendt, P.; Engelmann, M.; Knegendorf, L.; Vieyres, G.; Wedemeyer, H.; Hartmann, R.; Pietschmann, T.; et al. Antiviral activities of different interferon types and subtypes against hepatitis E virus replication. Antimicrob. Agents Chemother. 2016, 60, 2132–2139. [Google Scholar] [CrossRef] [Green Version]
  46. Wang, B.; Tian, D.; Sooryanarain, H.; Mahsoub, H.M.; Heffron, C.L.; Hassebroek, A.M.; Meng, X.J. Two mutations in the ORF1 of genotype 1 hepatitis E virus enhance virus replication and may associate with fulminant hepatic failure. Proc. Natl. Acad. Sci. USA 2022, 119, e2207503119. [Google Scholar] [CrossRef]
  47. Cierniak, F.; Ulrich, R.G.; Groschup, M.H.; Eiden, M. A modular hepatitis E virus replicon system for studies on the role of ORF1-encoded polyprotein domains. Pathogens 2022, 11, 355. [Google Scholar] [CrossRef] [PubMed]
  48. Graff, J.; Torian, U.; Nguyen, H.; Emerson, S.U. A bicistronic subgenomic mRNA encodes both the ORF2 and ORF3 proteins of hepatitis E virus. J. Virol. 2006, 80, 5919–5926. [Google Scholar] [CrossRef] [Green Version]
  49. Dao Thi, V.L.; Debing, Y.; Wu, X.; Rice, C.M.; Neyts, J.; Moradpour, D.; Gouttenoire, J. Sofosbuvir inhibits hepatitis E virus replication in vitro and results in an additive effect when combined with ribavirin. Gastroenterology 2016, 150, 82–85.e84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Szkolnicka, D.; Pollan, A.; Da Silva, N.; Oechslin, N.; Gouttenoire, J.; Moradpour, D. Recombinant hepatitis E viruses harboring tags in the ORF1 protein. J. Virol. 2019, 93, e00459-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Oechslin, N.; Da Silva, N.; Szkolnicka, D.; Cantrelle, F.X.; Hanoulle, X.; Moradpour, D.; Gouttenoire, J. Hepatitis E virus RNA-dependent RNA polymerase is involved in RNA replication and infectious particle production. Hepatology 2022, 75, 170–181. [Google Scholar] [CrossRef] [PubMed]
  52. Trozzi, C.; Bartholomew, L.; Ceccacci, A.; Biasiol, G.; Pacini, L.; Altamura, S.; Narjes, F.; Muraglia, E.; Paonessa, G.; Koch, U.; et al. In vitro selection and characterization of hepatitis C virus serine protease variants resistant to an active-site peptide inhibitor. J. Virol. 2003, 77, 3669–3679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xu, L.D.; Zhang, F.; Peng, L.; Luo, W.T.; Chen, C.; Xu, P.; Huang, Y.W. Stable Expression of a hepatitis E virus (HEV) RNA replicon in two mammalian cell lines to assess mechanism of innate immunity and antiviral response. Front. Microbiol. 2020, 11, 603699. [Google Scholar] [CrossRef] [PubMed]
  54. Nimgaonkar, I.; Archer, N.F.; Becher, I.; Shahrad, M.; LeDesma, R.A.; Mateus, A.; Caballero-Gomez, J.; Berneshawi, A.R.; Ding, Q.; Douam, F.; et al. Isocotoin suppresses hepatitis E virus replication through inhibition of heat shock protein 90. Antiviral Res. 2021, 185, 104997. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, C.; Freistaedter, A.; Schmelas, C.; Gunkel, M.; Dao Thi, V.L.; Grimm, D. An RNA interference/adeno-associated virus vector-based combinatorial gene therapy approach against hepatitis E virus. Hepatol. Commun. 2022, 6, 878–888. [Google Scholar] [CrossRef]
  56. Debing, Y.; Mishra, N.; Verbeken, E.; Ramaekers, K.; Dallmeier, K.; Neyts, J. A rat model for hepatitis E virus. Dis. Model. Mech. 2016, 9, 1203–1210. [Google Scholar] [CrossRef] [Green Version]
  57. Meister, T.L.; Bruggemann, Y.; Nocke, M.K.; Ulrich, R.G.; Schuhenn, J.; Sutter, K.; Gomer, A.; Bader, V.; Winklhofer, K.F.; Broering, R.; et al. A ribavirin-induced ORF2 single-nucleotide variant produces defective hepatitis E virus particles with immune decoy function. Proc. Natl. Acad. Sci. USA 2022, 119, e2202653119. [Google Scholar] [CrossRef]
  58. Ding, Q.; Heller, B.; Capuccino, J.M.; Song, B.; Nimgaonkar, I.; Hrebikova, G.; Contreras, J.E.; Ploss, A. Hepatitis E virus ORF3 is a functional ion channel required for release of infectious particles. Proc. Natl. Acad. Sci. USA 2017, 114, 1147–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Ding, Q.; Nimgaonkar, I.; Archer, N.F.; Bram, Y.; Heller, B.; Schwartz, R.E.; Ploss, A. Identification of the intragenomic promoter controlling hepatitis E virus subgenomic RNA transcription. mBio 2018, 9, e00769-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Todt, D.; Friesland, M.; Moeller, N.; Praditya, D.; Kinast, V.; Bruggemann, Y.; Knegendorf, L.; Burkard, T.; Steinmann, J.; Burm, R.; et al. Robust hepatitis E virus infection and transcriptional response in human hepatocytes. Proc. Natl. Acad. Sci. USA 2020, 117, 1731–1741. [Google Scholar] [CrossRef] [Green Version]
  61. Moradpour, D.; Evans, M.J.; Gosert, R.; Yuan, Z.; Blum, H.E.; Goff, S.P.; Lindenbach, B.D.; Rice, C.M. Insertion of green fluorescent protein into nonstructural protein 5A allows direct visualization of functional hepatitis C virus replication complexes. J. Virol. 2004, 78, 7400–7409. [Google Scholar] [CrossRef] [Green Version]
  62. Geng, J.; Fu, H.; Wang, L.; Bu, Q.; Liu, P.; Wang, M.; Sui, Y.; Wang, X.; Zhu, Y.; Zhuang, H. Phylogenetic analysis of the full genome of rabbit hepatitis E virus (rbHEV) and molecular biologic study on the possibility of cross species transmission of rbHEV. Infect. Genet. Evol. 2011, 11, 2020–2025. [Google Scholar] [CrossRef] [PubMed]
  63. Izopet, J.; Dubois, M.; Bertagnoli, S.; Lhomme, S.; Marchandeau, S.; Boucher, S.; Kamar, N.; Abravanel, F.; Guerin, J.L. Hepatitis E virus strains in rabbits and evidence of a closely related strain in humans, France. Emerg. Infect. Dis. 2012, 18, 1274–1281. [Google Scholar] [CrossRef] [PubMed]
  64. Sahli, R.; Fraga, M.; Semela, D.; Moradpour, D.; Gouttenoire, J. Rabbit HEV in immunosuppressed patients with hepatitis E acquired in Switzerland. J. Hepatol. 2019, 70, 1023–1025. [Google Scholar] [CrossRef] [Green Version]
  65. Lhomme, S.; Garrouste, C.; Kamar, N.; Saune, K.; Abravanel, F.; Mansuy, J.M.; Dubois, M.; Rostaing, L.; Izopet, J. Influence of polyproline region and macro domain genetic heterogeneity on HEV persistence in immunocompromised patients. J. Infect. Dis. 2014, 209, 300–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Munoz-Chimeno, M.; Cenalmor, A.; Garcia-Lugo, M.A.; Hernandez, M.; Rodriguez-Lazaro, D.; Avellon, A. Proline-rich hypervariable region of hepatitis E virus: Arranging the disorder. Microorganisms 2020, 8, 1417. [Google Scholar] [CrossRef] [PubMed]
  67. Todt, D.; Meister, T.L.; Steinmann, E. Hepatitis E virus treatment and ribavirin therapy: Viral mechanisms of nonresponse. Curr. Opin. Virol. 2018, 32, 80–87. [Google Scholar] [CrossRef] [PubMed]
  68. Lhomme, S.; Nicot, F.; Jeanne, N.; Dimeglio, C.; Roulet, A.; Lefebvre, C.; Carcenac, R.; Manno, M.; Dubois, M.; Peron, J.M.; et al. Insertions and duplications in the polyproline region of the hepatitis E virus. Front. Microbiol. 2020, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Shukla, P.; Nguyen, H.T.; Torian, U.; Engle, R.E.; Faulk, K.; Dalton, H.R.; Bendall, R.P.; Keane, F.E.; Purcell, R.H.; Emerson, S.U. Cross-species infections of cultured cells by hepatitis E virus and discovery of an infectious virus-host recombinant. Proc. Natl. Acad. Sci. USA 2011, 108, 2438–2443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Debing, Y.; Ramiere, C.; Dallmeier, K.; Piorkowski, G.; Trabaud, M.A.; Lebosse, F.; Scholtes, C.; Roche, M.; Legras-Lachuer, C.; de Lamballerie, X.; et al. Hepatitis E virus mutations associated with ribavirin treatment failure result in altered viral fitness and ribavirin sensitivity. J. Hepatol. 2016, 65, 499–508. [Google Scholar] [CrossRef]
  71. Papp, C.P.; Biedermann, P.; Harms, D.; Wang, B.; Kebelmann, M.; Choi, M.; Helmuth, J.; Corman, V.M.; Thurmer, A.; Altmann, B.; et al. Advanced sequencing approaches detected insertions of viral and human origin in the viral genome of chronic hepatitis E virus patients. Sci. Rep. 2022, 12, 1720. [Google Scholar] [CrossRef] [PubMed]
  72. Dalton, H.R.; Bendall, R.P.; Keane, F.E.; Tedder, R.S.; Ijaz, S. Persistent carriage of hepatitis E virus in patients with HIV infection. N. Engl. J. Med. 2009, 361, 1025–1027. [Google Scholar] [CrossRef] [PubMed]
  73. Metzger, K.; Bentaleb, C.; Hervouet, K.; Alexandre, V.; Montpellier, C.; Saliou, J.M.; Ferrie, M.; Camuzet, C.; Rouille, Y.; Lecoeur, C.; et al. Processing and subcellular localization of the hepatitis E virus replicase: Identification of candidate viral factories. Front. Microbiol. 2022, 13, 828636. [Google Scholar] [CrossRef]
  74. Nguyen, H.T.; Torian, U.; Faulk, K.; Mather, K.; Engle, R.E.; Thompson, E.; Bonkovsky, H.L.; Emerson, S.U. A naturally occurring human/hepatitis E recombinant virus predominates in serum but not in faeces of a chronic hepatitis E patient and has a growth advantage in cell culture. J. Gen. Virol. 2012, 93, 526–530. [Google Scholar] [CrossRef] [PubMed]
  75. Primadharsini, P.P.; Nagashima, S.; Nishiyama, T.; Takahashi, M.; Murata, K.; Okamoto, H. Development of recombinant infectious hepatitis E virus harboring the nanoKAZ gene and its application in drug screening. J. Virol. 2022, 96, e0190621. [Google Scholar] [CrossRef] [PubMed]
  76. Crooks, G.E.; Hon, G.; Chandonia, J.M.; Brenner, S.E. WebLogo: A sequence logo generator. Genome. Res. 2004, 14, 1188–1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Johne, R.; Reetz, J.; Ulrich, R.G.; Machnowska, P.; Sachsenroder, J.; Nickel, P.; Hofmann, J. An ORF1-rearranged hepatitis E virus derived from a chronically infected patient efficiently replicates in cell culture. J. Viral Hepat. 2014, 21, 447–456. [Google Scholar] [CrossRef] [PubMed]
  78. Takeuchi, S.; Yamazaki, Y.; Sato, K.; Takizawa, D.; Yamada, M.; Okamoto, H. Full genome analysis of a European-type genotype 3 hepatitis E virus variant obtained from a Japanese patient with autochthonous acute hepatitis E. J. Med. Virol. 2015, 87, 1067–1071. [Google Scholar] [CrossRef]
  79. Nishiyama, T.; Umezawa, K.; Yamada, K.; Takahashi, M.; Kunita, S.; Mulyanto; Kii, I.; Okamoto, H. The capsid (ORF2) protein of hepatitis E virus in feces is C-terminally truncated. Pathogens 2021, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  80. Dao Thi, V.L.; Wu, X.; Belote, R.L.; Andreo, U.; Takacs, C.N.; Fernandez, J.P.; Vale-Silva, L.A.; Prallet, S.; Decker, C.C.; Fu, R.M.; et al. Stem cell-derived polarized hepatocytes. Nat. Commun. 2020, 11, 1677. [Google Scholar] [CrossRef] [Green Version]
  81. Li, P.; Li, Y.; Wang, Y.; Liu, J.; Lavrijsen, M.; Li, Y.; Zhang, R.; Verstegen, M.M.A.; Wang, Y.; Li, T.C.; et al. Recapitulating hepatitis E virus-host interactions and facilitating antiviral drug discovery in human liver-derived organoids. Sci. Adv. 2022, 8, eabj5908. [Google Scholar] [CrossRef] [PubMed]
  82. Marceau, C.D.; Puschnik, A.S.; Majzoub, K.; Ooi, Y.S.; Brewer, S.M.; Fuchs, G.; Swaminathan, K.; Mata, M.A.; Elias, J.E.; Sarnow, P.; et al. Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens. Nature 2016, 535, 159–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schneider, W.M.; Luna, J.M.; Hoffmann, H.H.; Sanchez-Rivera, F.J.; Leal, A.A.; Ashbrook, A.W.; Le Pen, J.; Ricardo-Lax, I.; Michailidis, E.; Peace, A.; et al. Genome-scale identification of SARS-CoV-2 and pan-coronavirus host factor networks. Cell 2021, 184, 120–132.e114. [Google Scholar] [CrossRef]
  84. Shah, P.S.; Beesabathuni, N.S.; Fishburn, A.T.; Kenaston, M.W.; Minami, S.A.; Pham, O.H.; Tucker, I. Systems biology of virus-host protein interactions: From hypothesis generation to mechanisms of replication and pathogenesis. Annu. Rev. Virol. 2022, 9, 397–415. [Google Scholar] [CrossRef] [PubMed]
  85. Le, P.; Ahmed, N.; Yeo, G.W. Illuminating RNA biology through imaging. Nat. Cell Biol. 2022, 24, 815–824. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, J.; Liu, Y.; Wu, B.; Nikolaitchik, O.A.; Mohan, P.R.; Chen, J.; Pathak, V.K.; Hu, W.S. Visualizing the translation and packaging of HIV-1 full-length RNA. Proc. Natl. Acad. Sci. USA 2020, 117, 6145–6155. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Phylogenetic relationship of the Orthohepevirinae and genomic organization of hepatitis E virus (HEV). (A) Sequences of full-length genomes from various viruses of the Orthohepevirinae subfamily were aligned with MUSCLE, followed by phylogenetic tree building using the neighbor-joining method (Geneious Prime software, Biomatters). The name of the viral strain and the corresponding accession numbers are the following: HEV-1, Sar55 (AF444002); HEV-2, Mexican strain (HPENSSP); HEV-3k, HEV83-2-27 (AB740232); HEV-3b, pJE03-1760F (LC126332); HEV-3a, Kernow-C1 (JQ679014); HEV-3ra, rbIM223LR (LC484431); HEV-4, TW6196E (HQ634346); HEV-5, JBOAR135-Shiz09 (AB573435); HEV-6, wbJOY_06 (AB602441); HEV-7, 180C (KJ496144); HEV-8, M2 (MH410176); Paslahep. Alci, AlgSwe2012 (KF951328); Rocahep. ratti, LA-B350 (KM516906); Rocahep. eothenomi, KS_10_1641/GER/2009 (MK192405); Chirohep. rinolophi, HEV/Shanxi2013 (KJ562187); Chirohep eptesici, BtHEVMd2350 (KX513953); Chirohep. desmodi, DesRot/Peru/API17_F_DrHEV (MW249011); Avihep. magniiecur (AY535004); and Avihep. egretti, kocsag02/2014/HUN (KX589065). Sequences for which a molecular clone is available are indicated by a red dot. The scale bar indicates the number of nucleotide substitutions per site. (B) HEV possesses a 7.2-kb positive-strand RNA genome with a 5′ 7-methylguanylate cap (m7G cap) and a 3′ polyadenylated tail (poly-A) composed of, at least, three open reading frames (ORF). ORF1 encodes the viral replicase composed of several functional domains, including a methyltransferase (MeT), a putative papain-like cysteine protease (PCP), a hypervariable region (HVR), a macro domain, an RNA helicase (Hel), and an RNA-dependent RNA polymerase (RdRp). During RNA replication, a subgenomic RNA species is generated and gives rise to the expression of ORF2 and ORF3 proteins, respectively, the viral capsid, produced in different forms (not represented here), and a small protein involved in virus secretion.
Figure 1. Phylogenetic relationship of the Orthohepevirinae and genomic organization of hepatitis E virus (HEV). (A) Sequences of full-length genomes from various viruses of the Orthohepevirinae subfamily were aligned with MUSCLE, followed by phylogenetic tree building using the neighbor-joining method (Geneious Prime software, Biomatters). The name of the viral strain and the corresponding accession numbers are the following: HEV-1, Sar55 (AF444002); HEV-2, Mexican strain (HPENSSP); HEV-3k, HEV83-2-27 (AB740232); HEV-3b, pJE03-1760F (LC126332); HEV-3a, Kernow-C1 (JQ679014); HEV-3ra, rbIM223LR (LC484431); HEV-4, TW6196E (HQ634346); HEV-5, JBOAR135-Shiz09 (AB573435); HEV-6, wbJOY_06 (AB602441); HEV-7, 180C (KJ496144); HEV-8, M2 (MH410176); Paslahep. Alci, AlgSwe2012 (KF951328); Rocahep. ratti, LA-B350 (KM516906); Rocahep. eothenomi, KS_10_1641/GER/2009 (MK192405); Chirohep. rinolophi, HEV/Shanxi2013 (KJ562187); Chirohep eptesici, BtHEVMd2350 (KX513953); Chirohep. desmodi, DesRot/Peru/API17_F_DrHEV (MW249011); Avihep. magniiecur (AY535004); and Avihep. egretti, kocsag02/2014/HUN (KX589065). Sequences for which a molecular clone is available are indicated by a red dot. The scale bar indicates the number of nucleotide substitutions per site. (B) HEV possesses a 7.2-kb positive-strand RNA genome with a 5′ 7-methylguanylate cap (m7G cap) and a 3′ polyadenylated tail (poly-A) composed of, at least, three open reading frames (ORF). ORF1 encodes the viral replicase composed of several functional domains, including a methyltransferase (MeT), a putative papain-like cysteine protease (PCP), a hypervariable region (HVR), a macro domain, an RNA helicase (Hel), and an RNA-dependent RNA polymerase (RdRp). During RNA replication, a subgenomic RNA species is generated and gives rise to the expression of ORF2 and ORF3 proteins, respectively, the viral capsid, produced in different forms (not represented here), and a small protein involved in virus secretion.
Viruses 15 00869 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oechslin, N.; Ankavay, M.; Moradpour, D.; Gouttenoire, J. Expanding the Hepatitis E Virus Toolbox: Selectable Replicons and Recombinant Reporter Genomes. Viruses 2023, 15, 869. https://doi.org/10.3390/v15040869

AMA Style

Oechslin N, Ankavay M, Moradpour D, Gouttenoire J. Expanding the Hepatitis E Virus Toolbox: Selectable Replicons and Recombinant Reporter Genomes. Viruses. 2023; 15(4):869. https://doi.org/10.3390/v15040869

Chicago/Turabian Style

Oechslin, Noémie, Maliki Ankavay, Darius Moradpour, and Jérôme Gouttenoire. 2023. "Expanding the Hepatitis E Virus Toolbox: Selectable Replicons and Recombinant Reporter Genomes" Viruses 15, no. 4: 869. https://doi.org/10.3390/v15040869

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop