Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
PLoS Pathog ; 18(8): e1010798, 2022 08.
Article En | MEDLINE | ID: mdl-36007070

Hepatitis E virus (HEV) infection is the most common cause of acute viral hepatitis worldwide. Hepatitis E is usually asymptomatic and self-limiting but it can become chronic in immunocompromised patients and is associated with increased fulminant hepatic failure and mortality rates in pregnant women. HEV genome encodes three proteins including the ORF2 protein that is the viral capsid protein. Interestingly, HEV produces 3 isoforms of the ORF2 capsid protein which are partitioned in different subcellular compartments and perform distinct functions in the HEV lifecycle. Notably, the infectious ORF2 (ORF2i) protein is the structural component of virions, whereas the genome-free secreted and glycosylated ORF2 proteins likely act as a humoral immune decoy. Here, by using a series of ORF2 capsid protein mutants expressed in the infectious genotype 3 p6 HEV strain as well as chimeras between ORF2 and the CD4 glycoprotein, we demonstrated how an Arginine-Rich Motif (ARM) located in the ORF2 N-terminal region controls the fate and functions of ORF2 isoforms. We showed that the ARM controls ORF2 nuclear translocation likely to promote regulation of host antiviral responses. This motif also regulates the dual topology and functionality of ORF2 signal peptide, leading to the production of either cytosolic infectious ORF2i or reticular non-infectious glycosylated ORF2 forms. It serves as maturation site of glycosylated ORF2 by furin, and promotes ORF2-host cell membrane interactions. The identification of ORF2 ARM as a unique central regulator of the HEV lifecycle uncovers how viruses settle strategies to condense their genetic information and hijack cellular processes.


Hepatitis E virus , Hepatitis E , Amino Acid Motifs , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Female , Glycosylation , Hepatitis E/genetics , Hepatitis E/metabolism , Hepatitis E virus/growth & development , Humans , Pregnancy
2.
J Virol ; 95(23): e0058521, 2021 11 09.
Article En | MEDLINE | ID: mdl-34523963

Hepatitis E virus (HEV), an enterically transmitted RNA virus, is a major cause of acute hepatitis worldwide. Additionally, HEV genotype 3 (gt3) can frequently persist in immunocompromised individuals with an increased risk for developing severe liver disease. Currently, no HEV-specific treatment is available. The viral open reading frame 3 (ORF3) protein facilitates HEV egress in vitro and is essential for establishing productive infection in macaques. Thus, ORF3, which is unique to HEV, has the potential to be explored as a target for antiviral therapy. However, significant gaps exist in our understanding of the critical functions of ORF3 in HEV infection in vivo. Here, we utilized a polarized hepatocyte culture model and a human liver chimeric mouse model to dissect the roles of ORF3 in gt3 HEV release and persistent infection. We show that ORF3's absence substantially decreased HEV replication and virion release from the apical surface but not the basolateral surface of polarized hepatocytes. While wild-type HEV established a persistent infection in humanized mice, mutant HEV lacking ORF3 (ORF3null) failed to sustain the infection despite transient replication in the liver and was ultimately cleared. Strikingly, mice inoculated with the ORF3null virus displayed no fecal shedding throughout the 6-week experiment. Overall, our results demonstrate that ORF3 is required for HEV fecal shedding and persistent infection, providing a rationale for targeting ORF3 as a treatment strategy for HEV infection. IMPORTANCE HEV infections are associated with significant morbidity and mortality. HEV gt3 additionally can cause persistent infection, which can rapidly progress to liver cirrhosis. Currently, no HEV-specific treatments are available. The poorly understood HEV life cycle hampers the development of antivirals for HEV. Here, we investigated the role of the viral ORF3 protein in HEV infection in polarized hepatocyte cultures and human liver chimeric mice. We found that two major aspects of the HEV life cycle require ORF3: fecal virus shedding and persistent infection. These results provide a rationale for targeting ORF3 to treat HEV infection.


Hepatitis E virus/growth & development , Hepatitis E virus/genetics , Hepatitis E/virology , Hepatocytes/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Release , Animals , Antiviral Agents/pharmacology , Liver , Mice , Open Reading Frames , Persistent Infection , Virion , Virus Replication
3.
Viruses ; 13(3)2021 03 04.
Article En | MEDLINE | ID: mdl-33806591

Hepatitis E virus (HEV) is considered as an emerging global health problem. In most cases, hepatitis E is a self-limiting disease and the virus is cleared spontaneously without the need of antiviral therapy. However, immunocompromised individuals can develop chronic infection and liver fibrosis that can progress rapidly to cirrhosis and liver failure. The lack of efficient and relevant cell culture system and animal models has limited our understanding of the biology of HEV and the development of effective drugs for chronic cases. In the present study, we developed a model of persistent HEV infection in human hepatocytes in which HEV replicates efficiently. This HEV cell culture system is based on differentiated HepaRG cells infected with an isolate of HEV-3 derived from a patient suffering from acute hepatitis E. Efficient replication was maintained for several weeks to several months as well as after seven successive passages on HepaRG naïve cells. Moreover, after six passages onto HepaRG, we found that the virus was still infectious after oral inoculation into pigs. We also showed that ribavirin had an inhibitory effect on HEV replication in HepaRG. In conclusion, this system represents a relevant and efficient in vitro model of HEV replication that could be useful to study HEV biology and identify effective antiviral drugs against chronic HEV infection.


Cell Culture Techniques/methods , Hepatitis E virus/growth & development , Hepatitis E/virology , Hepatocytes/virology , Animals , Cell Line , Hepatocytes/cytology , Humans , Swine , Virus Replication
4.
Viruses ; 12(10)2020 10 09.
Article En | MEDLINE | ID: mdl-33050353

Hepatitis E virus (HEV) (family Hepeviridae) is one of the most common human pathogens, causing acute hepatitis and an increasingly recognized etiological agent in chronic hepatitis and extrahepatic manifestations. Recent studies reported that not only are the classical members of the species Orthohepevirus A (HEV-A) pathogenic to humans but a genetically highly divergent rat origin hepevirus (HEV-C1) in species Orthohepevirus C (HEV-C) is also able to cause zoonotic infection and symptomatic disease (hepatitis) in humans. This review summarizes the current knowledge of hepeviruses in rodents with special focus of rat origin HEV-C1. Cross-species transmission and genetic diversity of HEV-C1 and confirmation of HEV-C1 infections and symptomatic disease in humans re-opened the long-lasting and full of surprises story of HEV in human. This novel knowledge has a consequence to the epidemiology, clinical aspects, laboratory diagnosis, and prevention of HEV infection in humans.


Disease Reservoirs/veterinary , Hepatitis E virus/growth & development , Hepatitis E/transmission , Hepatitis, Viral, Animal/transmission , Animals , Cell Line , Disease Reservoirs/virology , Genome, Viral/genetics , Hepatitis E virus/genetics , Hepevirus/growth & development , Humans , Phylogeny , Rats , Zoonoses/transmission , Zoonoses/virology
5.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Article En | MEDLINE | ID: mdl-32374474

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Cytokines/genetics , Dendritic Cells/drug effects , Immunologic Factors/pharmacology , Macrophages/drug effects , Triterpenes/pharmacology , Betacoronavirus/growth & development , Betacoronavirus/immunology , Cell Differentiation/drug effects , Chikungunya virus/drug effects , Chikungunya virus/growth & development , Chikungunya virus/immunology , Cytokines/classification , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Ebolavirus/drug effects , Ebolavirus/growth & development , Ebolavirus/immunology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepatitis E virus/drug effects , Hepatitis E virus/growth & development , Hepatitis E virus/immunology , Humans , Immunity, Innate/drug effects , Macrophages/immunology , Macrophages/virology , Organ Specificity , Picornaviridae/drug effects , Picornaviridae/growth & development , Picornaviridae/immunology , Primary Cell Culture , SARS-CoV-2 , Signal Transduction , Zika Virus/drug effects , Zika Virus/growth & development , Zika Virus/immunology
6.
Int J Food Microbiol ; 325: 108625, 2020 Jul 16.
Article En | MEDLINE | ID: mdl-32361052

Infection with the hepatitis E virus (HEV) can cause acute and chronic hepatitis in humans. The zoonotic HEV genotype 3 is mainly transmitted by consumption of raw and fermented meat products prepared from infected pigs or wild boars. Lowering of pH during fermentation is one of the microbiological hurdles considered to inhibit growth of certain pathogens. However, no data are currently available on pH stability of HEV. As a reliable and reproducible measurement of HEV infectivity in meat products is not established so far, the stability of the cell culture-adapted HEV genotype 3 strain 47832c was analyzed here in phosphate-buffered saline (PBS) at different pH values. Only a minimal decrease of infectivity (up to 0.6 log10 focus forming units) was found after treatment at pH 2 to 9 for 3 h at room temperature. At pH 10, a decrease of about 3 log10 was evident, whereas no remaining virus (>3.5 log10 decrease) was detected at pH 1. The conditions usually achieved during curing of raw sausages were simulated using D/L-lactic acid added to PBS resulting in pH 4.5 to 6.5. After incubation at 4 °C for 7 days at these conditions, no significant differences as compared to a standard PBS solution at pH 7.7 were evident. At room temperature, a 0.8 log10 decrease was found at pH 4.7 after 7 days incubation compared to pH 7.7, but less at the other pH values. In conclusion, only minimal inactivating effects were found at pH conditions commonly occurring during food processing. Therefore, remaining infectious virus might be present in fermented meat products if HEV-contaminated starting material was used. Additional effects of other factors like high salt concentrations and low aw values should be investigated in future studies.


Hepatitis E virus/growth & development , Lactic Acid/pharmacology , Meat Products/virology , Virus Inactivation/drug effects , Animals , Hepatitis E/virology , Hepatitis E virus/genetics , Humans , Hydrogen-Ion Concentration , Sus scrofa , Swine
7.
Sci Rep ; 10(1): 2753, 2020 02 17.
Article En | MEDLINE | ID: mdl-32066795

Hepatitis E virus (HEV) infection contributes to a considerable proportion of acute-on-chronic liver failure (ACLF) in patients with chronic hepatitis B virus (HBV) infection. This study aimed to predict the prognosis of chronic HBV infection patients precipitating acute HEV infection. A total of 193 patients were enrolled in this study. The performances of three chronic liver disease prognostic models (CTP score, MELD score, and CLIF-C ADs) were analyzed for predicting the development of ACLF following HEV superimposing chronic HBV infection. Subsequently, the performances of five ACLF prognostic assessment models (CTP score, MELD score, CLIF-C ACLFs, CLIF-C OFs, and COSSH-ACLFs) were analyzed for predicting the outcome of those ACLF patients. Of 193 chronic HBV infection patients precipitating acute HEV infection, 13 patients were diagnosed ACLF on admission, 54 patients developed to ACLF after admission, and 126 patients had non-ACLF during the stay in hospital. For predicting the development of ACLF, CTP score yielded a significantly higher AUROC compared with MELD score and CLIF-C ADs (0.92, 0.88, and 0.86, respectively; all p < 0.05). For predicting the poor prognosis of ACLF patients, the COSSH-ACLFs yielded a significantly higher AUROC compared with CLIF-C ACLFs, CLIF-C OFs, MELD score, and CTP score (0.89, 0.83, 0.81, 0.67, and 0.58, respectively; all p < 0.05). In conclusion, the stepwise application of CTP score and COSSH-ACLFs can predict the prognosis of chronic HBV infection patients precipitating acute HEV infection.


Acute-On-Chronic Liver Failure/diagnosis , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/diagnosis , Hepatitis E virus/pathogenicity , Hepatitis E/diagnosis , Acute Disease , Acute-On-Chronic Liver Failure/mortality , Acute-On-Chronic Liver Failure/pathology , Acute-On-Chronic Liver Failure/virology , Adult , Aged , Bilirubin/blood , Biomarkers/blood , Coinfection , Creatinine/blood , Female , Hepatitis B virus/growth & development , Hepatitis B, Chronic/mortality , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Hepatitis E/mortality , Hepatitis E/pathology , Hepatitis E/virology , Hepatitis E virus/growth & development , Humans , Liver Function Tests , Liver Transplantation , Male , Middle Aged , Prognosis , ROC Curve , Retrospective Studies , Survival Analysis
8.
Viruses ; 12(2)2020 Jan 24.
Article En | MEDLINE | ID: mdl-31991673

Hepatitis E virus (HEV) is a major concern in public health worldwide. Infections with HEV genotypes 3, 4, or 7 can lead to chronic hepatitis while genotype 1 infections can trigger severe hepatitis in pregnant women. Infections with all genotypes can worsen chronic liver diseases. As virions are lipid-associated in blood and naked in feces, efficient methods of propagating HEV clinical strains in vitro and evaluating the infectivity of both HEV forms are needed. We evaluated the spread of clinical strains of HEV genotypes 1 (HEV1) and 3 (HEV3) by quantifying viral RNA in culture supernatants and cell lysates. Infectivity was determined by endpoint dilution and calculation of the tissue culture infectious dose 50 (TCID50). An enhanced HEV production could be obtained varying the composition of the medium, including fetal bovine serum (FBS) and dimethylsulfoxide (DMSO) content. This increased TCID50 from 10 to 100-fold and allowed us to quantify HEV1 infectivity. These optimized methods for propagating and measuring HEV infectivity could be applied to health safety processes and will be useful for testing new antiviral drugs.


Hepatitis E virus/growth & development , Virus Cultivation/methods , Culture Media , Genotype , Hep G2 Cells , Hepatitis E virus/genetics , Humans , RNA, Viral/analysis
9.
J Virol ; 93(19)2019 10 01.
Article En | MEDLINE | ID: mdl-31315997

Hepatitis E virus (HEV) is one of the most common causes of acute hepatitis and jaundice in the world. Current understanding of the molecular virology and pathogenesis of hepatitis E is incomplete, due particularly to the limited availability of functional tools. Here, we report the development of tagged HEV genomes as a novel tool to investigate the viral life cycle. A selectable subgenomic HEV replicon was subjected to random 15-nucleotide sequence insertion using transposon-based technology. Viable insertions in the open reading frame 1 (ORF1) protein were selected in a hepatoblastoma cell line. Functional insertion sites were identified downstream of the methyltransferase domain, in the hypervariable region (HVR), and between the helicase and RNA-dependent RNA polymerase domains. HEV genomes harboring a hemagglutinin (HA) epitope tag or a small luciferase (NanoLuc) in the HVR were found to be fully functional and to allow the production of infectious virus. NanoLuc allowed quantitative monitoring of HEV infection and replication by luciferase assay. The use of HA-tagged replicons and full-length genomes allowed localization of putative sites of HEV RNA replication by the simultaneous detection of viral RNA by fluorescence in situ hybridization and of ORF1 protein by immunofluorescence. Candidate HEV replication complexes were found in cytoplasmic dot-like structures which partially overlapped ORF2 and ORF3 proteins as well as exosomal markers. Hence, tagged HEV genomes yield new insights into the viral life cycle and should allow further investigation of the structure and composition of the viral replication complex.IMPORTANCE Hepatitis E virus (HEV) infection is an important cause of acute hepatitis and may lead to chronic infection in immunocompromised patients. Knowledge of the viral life cycle is incomplete due to the limited availability of functional tools. In particular, low levels of expression of the ORF1 protein or limited sensitivity of currently available antibodies or both limit our understanding of the viral replicase. Here, we report the successful establishment of subgenomic HEV replicons and full-length genomes harboring an epitope tag or a functional reporter in the ORF1 protein. These novel tools should allow further characterization of the HEV replication complex and to improve our understanding of the viral life cycle.


Hemagglutinins/metabolism , Hepatitis E virus/growth & development , Proteins/metabolism , Recombinant Proteins/metabolism , Staining and Labeling/methods , Cell Line, Tumor , DNA Transposable Elements , Hemagglutinins/genetics , Hepatitis E virus/genetics , Hepatocytes/virology , Humans , Mutagenesis, Insertional , Proteins/genetics , Recombinant Proteins/genetics , Virus Replication
10.
Viruses ; 11(7)2019 07 03.
Article En | MEDLINE | ID: mdl-31277308

Despite a growing awareness, hepatitis E virus (HEV) remains understudied and investigations have been historically hampered by the absence of efficient cell culture systems. As a result, the pathogenesis of HEV infection and basic steps of the HEV life cycle are poorly understood. Major efforts have recently been made through the development of HEV infectious clones and cellular systems that significantly advanced HEV research. Here, we summarize these systems, discussing their advantages and disadvantages for HEV studies. We further capitalize on the need for HEV-permissive polarized cell models to better recapitulate the entire HEV life cycle and transmission.


Cell Culture Techniques/methods , Hepatitis E virus/growth & development , Carcinoma, Hepatocellular , Cell Line , Hepatitis E/virology , Hepatocytes/virology , Humans , Life Cycle Stages/physiology , Stem Cells
11.
Antiviral Res ; 170: 104570, 2019 10.
Article En | MEDLINE | ID: mdl-31362004

Hepatitis E is a global public health problem. Ribavirin (RBV) and pegylated interferon alpha are currently administered to cure hepatitis E. Recently, in combination with RBV, sofosbuvir (SOF), an anti-hepatitis C virus nucleotide analog, is also given to patients with chronic hepatitis E. However, this combinatorial therapy sometimes fails to achieve a sustained virological response. In this study, we used 27 antiviral compounds, including 15 nucleos(t)ide analogs, for in vitro screening against a genotype 3 HEV strain containing a Gaussia luciferase reporter. RBV, SOF, 2'-C-methyladenosine, 2'-C-methylcytidine (2CMC), 2'-C-methylguanosine (2CMG), and two 4'-azido nucleoside analogs (R-1479 and RO-9187) suppressed replication of the reporter genome, while only RBV, SOF, 2CMC and 2CMG inhibited the growth of genotype 3 HEV in cultured cells. Although 2CMG and RBV (2CMG/RBV) exhibited a synergistic effect while SOF/RBV and 2CMC/RBV showed antagonistic effects on the reporter assay, these three nucleos(t)ide analogs acted additively with RBV in inhibiting HEV growth in cultured cells. Furthermore, SOF and 2CMG, with four interferons (IFN-α2b, IFN-λ1, IFN-λ2 and IFN-λ3), inhibited HEV growth efficiently and cleared HEV in cultured cells. These results suggest that, in combination with RBV or interferons, SOF and 2CMG would be promising bases for developing anti-HEV nucleos(t)ide analogs.


Antiviral Agents/pharmacology , Hepatitis E virus/drug effects , Hepatitis E virus/growth & development , Nucleosides/pharmacology , Cell Line , Cell Survival/drug effects , Drug Discovery , Drug Synergism , Genes, Reporter , Genotype , Hepatitis E/drug therapy , Hepatitis E/virology , Hepatitis E virus/genetics , Humans , Interferons/pharmacology , Luciferases , Nucleosides/chemistry
12.
Viruses ; 11(6)2019 06 18.
Article En | MEDLINE | ID: mdl-31216711

Hepatitis E virus (HEV) is an underdiagnosed pathogen with approximately 20 million infections each year and currently the most common cause of acute viral hepatitis. HEV was long considered to be confined to developing countries but there is increasing evidence that it is also a medical problem in the Western world. HEV that infects humans belongs to the Orthohepevirus A species of the Hepeviridae family. Novel HEV-like viruses have been observed in a variety of animals and some have been shown to be able to cross the species barrier, causing infection in humans. Several cell culture models for HEV have been established in the past years, but their efficiency is usually relatively low. With the circulation of this virus and related viruses in a variety of species, several different animal models have been developed. In this review, we give an overview of these animal models, indicate their main characteristics, and highlight how they may contribute to our understanding of the basic aspects of the viral life cycle and cross-species infection, the study of pathogenesis, and the evaluation of novel preventative and therapeutic strategies.


Disease Models, Animal , Hepatitis E virus/growth & development , Hepatitis E/pathology , Animals , Hepatitis E/physiopathology , Hepatitis E virus/pathogenicity
13.
Antiviral Res ; 163: 34-49, 2019 03.
Article En | MEDLINE | ID: mdl-30653997

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and is the leading cause of enterically-transmitted viral hepatitis worldwide. Increasing numbers of HEV infections, together with no available specific anti-HEV treatment, contributes to the pathogen's major health burden. A robust cell culture system is required for virologic studies and the development of new antiviral drugs. Unfortunately, like other hepatitis viruses, HEV is difficult to propagate in conventional cell lines. Many different cell culture systems have been tested using various HEV strains, but viral replication usually progresses very slowly, and infection with low virion counts results in non-productive HEV replication. However, recent progress involving generation of cDNA clones and passaging primary patient isolates in distinct cell lines has improved in vitro HEV propagation. This review describes various approaches to cultivate HEV in cellular and animal models and how these systems are used to study HEV infections and evaluate anti-HEV drug candidates.


Cell Culture Techniques/methods , Hepatitis E virus/growth & development , Virus Cultivation/methods , Animals , Cell Line , Disease Models, Animal , Hepatitis E/virology , Humans
14.
Article En | MEDLINE | ID: mdl-29735581

Novel hepeviruses have been recovered from many different animal species in recent years, increasing the diversity known to exist among the Hepeviridae, which now include two genera, Piscihepevirus and Orthohepevirus Multiple viral genotypes in the Orthohepevirus A species are able to replicate and cause acute hepatitis E in humans, and thus represent an important public health problem in industrialized as well as developing countries. Although hepatitis E virus (HEV) infections typically result in acute and self-limited hepatitis, immunocompromised and transplant patients are vulnerable to prolonged infections and to chronic hepatitis. Cell culture systems have been established for several HEV strains and offer new opportunities for the study of HEV biology. Similarly, a variety of new small animal models have been developed, using either nonhuman hepeviruses in their cognate hosts as surrogates for human HEV, or human HEV infection of immunodeficient mice with chimeric livers engrafted with human hepatocytes. These new models provide several advantages over previous nonhuman primate models of hepatitis E infection and will facilitate studies of pathogenicity, cross-species infection, mechanisms of virus replication, and vaccine and antiviral agent development. This article reviews the current understanding of small animal models for HEV.


Disease Models, Animal , Hepatitis E virus/growth & development , Hepatitis E/pathology , Animals , Antiviral Agents/therapeutic use , Chickens , Ferrets , Gerbillinae , Hepatitis E/drug therapy , Hepatitis E/prevention & control , Hepatitis E virus/pathogenicity , Mice , Rabbits , Rats , Tupaiidae , Viral Hepatitis Vaccines/immunology
15.
Virology ; 527: 116-121, 2019 01 15.
Article En | MEDLINE | ID: mdl-30496912

Hepatitis E virus (HEV) infection is widespread in the global pig population. Although clinically inapparent in pigs, HEV infection is the cause of Hepatitis E in humans and transmission via the food chain has been established. Following a 2013 study that investigated prevalence of HEV infection in UK slaughter-age pigs samples indicating highest viral load were selected for further characterisation. High throughput sequencing was used to obtain the complete coding sequence from five samples. An in-frame insertion was observed within the HEV hypervariable region in two samples. To interrogate whether this mutation may be the cause of high-level viraemia and faecal shedding as observed in the sampled pigs virus isolation and culture was conducted. Based on viral growth kinetics there was no evidence that these insertions affected replication efficiency in vitro, suggesting as yet undetermined host factors may affect the course of infection and consequently the risk of foodborne transmission.


Hepatitis E virus/genetics , Hepatitis E/veterinary , Sus scrofa/virology , Viremia/veterinary , Animals , Feces/virology , Food Microbiology , Genome, Viral/genetics , Hepatitis E/epidemiology , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/growth & development , Mutagenesis, Insertional , Open Reading Frames , Phylogeny , Prevalence , RNA, Viral/genetics , Sequence Analysis, RNA , Swine , United Kingdom/epidemiology , Viremia/virology
16.
Antiviral Res ; 157: 151-158, 2018 09.
Article En | MEDLINE | ID: mdl-30036559

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and a member of the genus Orthohepevirus in the family Hepeviridae. HEV infections are the common cause of acute hepatitis but can also take chronic courses. Ribavirin is the treatment of choice for most patients and type I interferon (IFN) has been evaluated in a few infected transplantation patients in vivo. However, no effective and specific treatments against HEV infections are currently available. In this study, we evaluated the natural compound silvestrol, isolated from the plant Aglaia foveolata, and known for its specific inhibition of the DEAD-box RNA helicase eIF4A in state-of-the-art HEV experimental model systems. Silvestrol blocked HEV replication of different subgenomic replicons in a dose-dependent manner at low nanomolar concentrations and acted additive to ribavirin (RBV). In addition, HEV p6-based full length replication and production of infectious particles was reduced in the presence of silvestrol. A pangenotypic effect of the compound was further demonstrated with primary isolates from four different human genotypes in HEV infection experiments of hepatocyte-like cells derived from human embryonic and induced pluripotent stem cells. In vivo, HEV RNA levels rapidly declined in the feces of treated mice while no effect was observed in the vehicle treated control animals. In conclusion, silvestrol could be identified as pangenotypic HEV replication inhibitor in vitro with additive effect to RBV and further demonstrated high potency in vivo. The compound therefore may be considered in future treatment strategies of chronic hepatitis E in immunocompromised patients.


Antiviral Agents/pharmacology , Hepatitis E virus/drug effects , Hepatitis E/drug therapy , Triterpenes/pharmacology , Virus Replication/drug effects , Aglaia/chemistry , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/isolation & purification , Cells, Cultured , Disease Models, Animal , Drug Interactions , Feces/virology , Hepatitis E virus/growth & development , Humans , Mice , Ribavirin/pharmacology , Triterpenes/administration & dosage , Triterpenes/isolation & purification , Viral Load
17.
DNA Cell Biol ; 37(7): 593-599, 2018 Jul.
Article En | MEDLINE | ID: mdl-29897788

Hepatitis E virus (HEV) is a major cause of viral hepatitis worldwide. Owing to its feco oral transmission route, sporadic as well as epidemic outbreaks recurrently occur. No specific antiviral therapy is available against the disease caused by HEV. Broad spectrum antivirals such as ribavirin and interferon alfa are prescribed in severe and chronic HEV cases. However, the side effects, cost, and limitations of usage render the available treatment unsuitable for several categories of patients. We recently reported the ability of zinc to inhibit viral replication in mammalian cell culture models of HEV infection. Zinc will be a safe and economical antiviral therapy option if it inhibits HEV replication during the natural course of infection. This essay discusses the putative mechanism(s) by which zinc inhibits HEV replication and provides an overview of the possible therapeutic potential of zinc in HEV patients.


Antiviral Agents/pharmacology , Hepatitis E virus/drug effects , Hepatitis E/drug therapy , Virus Replication/drug effects , Zinc Compounds/pharmacology , Animals , Gene Expression Regulation , Hepatitis E/genetics , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/growth & development , Hepatitis E virus/metabolism , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/drug effects , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferons , Interleukins/genetics , Interleukins/immunology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Signal Transduction , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
18.
Food Environ Virol ; 10(3): 225-252, 2018 09.
Article En | MEDLINE | ID: mdl-29623595

Historically in developed countries, reported hepatitis E cases were typically travellers returning from countries where hepatitis E virus (HEV) is endemic, but now there are increasing numbers of non-travel-related ("autochthonous") cases being reported. Data for HEV in New Zealand remain limited and the transmission routes unproven. We critically reviewed the scientific evidence supporting HEV transmission routes in other developed countries to inform how people in New Zealand may be exposed to this virus. A substantial body of indirect evidence shows domesticated pigs are a source of zoonotic human HEV infection, but there is an information bias towards this established reservoir. The increasing range of animals in which HEV has been detected makes it important to consider other possible animal reservoirs of HEV genotypes that can or could infect humans. Foodborne transmission of HEV from swine and deer products has been proven, and a large body of indirect evidence (e.g. food surveys, epidemiological studies and phylogenetic analyses) support pig products as vehicles of HEV infection. Scarce data from other foods suggest we are neglecting other potential sources of foodborne HEV infection. Moreover, other transmission routes are scarcely investigated in developed countries; the role of infected food handlers, person-to-person transmission via the faecal-oral route, and waterborne transmission from recreational contact or drinking untreated or inadequately treated water. People have become symptomatic after receiving transfusions of HEV-contaminated blood, but it is unclear how important this is in the overall hepatitis E disease burden. There is need for broader research efforts to support establishing risk-based controls.


Developed Countries , Disease Transmission, Infectious , Foodborne Diseases/virology , Hepatitis E virus/growth & development , Hepatitis E/transmission , Waterborne Diseases/virology , Zoonoses/virology , Animal Diseases/virology , Animals , Deer/virology , Drinking Water/virology , Feces/virology , Hepatitis E/virology , Humans , Meat/virology , New Zealand , Swine/virology
19.
J Virol ; 92(11)2018 06 01.
Article En | MEDLINE | ID: mdl-29540601

The molecular mechanisms of liver pathology and clinical disease in hepatitis E virus (HEV) infection remain unclear. MicroRNAs (miRNAs) are known to modulate viral pathogenesis either by directly altering viral gene expression or by enhancing cellular antiviral responses. Given the importance of microRNA-122 (miR-122) in liver pathobiology, we investigated possible role of miR-122 in HEV infection. In silico predictions using HEV genotype 1 (HEV-1), HEV-2, HEV-3, and HEV-4 sequences showed that the majority of genomes (203/222) harbor at least one miR-122/microRNA-122-3p (miR-122*) target site. Interestingly, HEV-1 genomes showed a highly (97%) conserved miR-122 target site in the RNA-dependent RNA polymerase (RdRp) region (RdRpc). We analyzed the significance of miR-122 target sites in HEV-1/HEV-3 (HEV-1/3) genomes by using a replicon-based cell culture system. HEV infection did not change the basal levels of miR-122 in hepatoma cells. However, transfection of these cells with miR-122 mimics enhanced HEV-1/3 replication and depletion of miR-122 with inhibitors led to suppression of HEV-1/3 replication. Mutant HEV-1 replicons with an altered target RdRpc sequence (CACTCC) showed a drastic decrease in virus replication, whereas introduction of alternative miR-122 target sites in mutant replicons rescued viral replication. There was enrichment of HEV-1 RNA and miR-122 molecules in RNA-induced silencing complexes in HEV-infected cells. Furthermore, pulldown of miR-122 molecules from HEV-infected cells resulted in pulldown of HEV genomic RNA along with miR-122 molecules. These observations indicate that miR-122 facilitates HEV-1 replication, probably via direct interaction with a target site in the viral genome. The positive role of miR-122 in viral replication presents novel opportunities for antiviral therapy and management of hepatitis E.IMPORTANCE Hepatitis E is a problem in both developing and developed countries. HEV infection in most patients follows a self-limited course; however, 20% to 30% mortality is seen in infected pregnant women. HEV superinfections in patients with chronic hepatitis B or hepatitis C virus infections are associated with adverse clinical outcomes, and both conditions warrant therapy. Chronic HEV infections in immunocompromised transplant recipients are known to rapidly progress into cirrhosis. Currently, off-label use of ribavirin (RBV) and polyethylene glycol-interferon (PEG-IFN) as antiviral therapy has shown promising results in both acute and chronic hepatitis E patients; however, the teratogenicity of RBV limits its use during pregnancy, while alpha IFN (IFN-α) increases the risk of transplant rejections. Experimental data determined with genotype 1 virus in the current study show that miR-122 facilitates HEV replication. These observations present novel opportunities for antiviral therapy and management of hepatitis E.


Hepatitis E virus/growth & development , Hepatitis E virus/genetics , Hepatitis E/therapy , MicroRNAs/genetics , Virus Replication/genetics , A549 Cells , Argonaute Proteins/metabolism , Base Sequence , Cell Line, Tumor , Genome, Viral/genetics , Hep G2 Cells , Hepatitis E/pathology , Hepatitis E/virology , Hepatitis E virus/classification , Humans , Liver/pathology , Liver/virology , RNA-Dependent RNA Polymerase/genetics
20.
Curr Protoc Microbiol ; 48: 15L.1.1-15L.1.15, 2018 02 22.
Article En | MEDLINE | ID: mdl-29512115

Hepatitis E virus (HEV) predominantly causes acute liver disease in humans and is transmitted via the fecal-oral route. HEV infection in pregnant women can result in grave consequences, with up to 30% fatality. The HEV strains infecting humans mainly belong to four genotypes. Genotypes 1 and 2 are restricted to human infection, while genotypes 3 and 4 are zoonotic. HEV genotype 3 (HEV-3) can cause both acute and chronic liver diseases. Several cell lines (mainly hepatocytes) have been developed for HEV propagation and biological study. However, HEV production in these cell lines is suboptimal and inefficient. Here, we present methods for the isolation, propagation, and quantification of HEV. © 2018 by John Wiley & Sons, Inc.


Cell Culture Techniques/methods , Hepatitis E virus/isolation & purification , Hepatitis E/virology , Polymerase Chain Reaction/methods , Virus Cultivation/methods , Feces/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Hepatitis E virus/growth & development , Humans
...