Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Hepatology ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776183
2.
Nat Rev Dis Primers ; 9(1): 51, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37770459

ABSTRACT

Hepatitis A is a vaccine-preventable infection caused by the hepatitis A virus (HAV). Over 150 million new infections of hepatitis A occur annually. HAV causes an acute inflammatory reaction in the liver that usually resolves spontaneously without chronic sequelae. However, up to 20% of patients experience a prolonged or relapsed course and <1% experience acute liver failure. Host factors, such as immunological status, age, pregnancy and underlying hepatic diseases, can affect the severity of disease. Anti-HAV IgG antibodies produced in response to HAV infection persist for life and protect against re-infection; vaccine-induced antibodies against hepatitis A confer long-term protection. The WHO recommends vaccination for individuals at higher risk of infection and/or severe disease in countries with very low and low hepatitis A virus endemicity, and universal childhood vaccination in intermediate endemicity countries. To date, >25 countries worldwide have implemented such programmes, resulting in a reduction in the incidence of HAV infection. Improving hygiene and sanitation, rapid identification of outbreaks and fast and accurate intervention in outbreak control are essential to reducing HAV transmission.

3.
Nat Rev Microbiol ; 21(9): 573-589, 2023 09.
Article in English | MEDLINE | ID: mdl-37185947

ABSTRACT

Infectious hepatitis type A and type E are caused by phylogenetically distinct single-stranded, positive-sense RNA viruses that were once considered to be non-enveloped. However, studies show that both are released nonlytically from hepatocytes as 'quasi-enveloped' virions cloaked in host membranes. These virion types predominate in the blood of infected individuals and mediate virus spread within the liver. They lack virally encoded proteins on their surface and are resistant to neutralizing anti-capsid antibodies induced by infection, yet they efficiently enter cells and initiate new rounds of virus replication. In this Review, we discuss the mechanisms by which specific peptide sequences in the capsids of these quasi-enveloped virions mediate their endosomal sorting complexes required for transport (ESCRT)-dependent release from hepatocytes through multivesicular endosomes, what is known about how they enter cells, and the impact of capsid quasi-envelopment on host immunity and pathogenesis.


Subject(s)
Liver , Virus Internalization , Humans , Capsid Proteins , Capsid/metabolism , Hepatitis Viruses/metabolism , Virion/metabolism
4.
Hepatology ; 78(6): 1867-1881, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37185268

ABSTRACT

BACKGROUND AND AIMS: The HEV is a small positive-sense RNA virus that encodes a cytoplasmic form of the capsid protein (ORF2c), essential for virion structure, and a secreted glycosylated form (ORF2s) that accumulates at high titer in serum and can mask neutralizing epitopes. We explored the contribution of ORF2s to HEV replication and its role in generating antibodies against ORF2 in a nonhuman primate model. APPROACH AND RESULTS: We used a recombinant HEV genotype 3 variant that does not express ORF2s due to the introduction of stop codons (ORF2s mut ). Rhesus macaques (RMs) were given intrahepatic injections of infectious wildtype HEV (ORF2s wt ) RNA or a variant lacking ORF2s expression (ORF2s mut ). The replication of the ORF2s mut virus was delayed by ~2 weeks compared with ORF2s wt , and peak titers were nearly tenfold lower. Reversions of the 3 mutations that blocked ORF2s expression were not detected in the ORF2s mut genomes, indicating genetic stability. However, serum antibodies against ORF2 were transiently detected in RMs infected with ORF2s mut , whereas they were long-lasting in RMs infected with ORF2s wt . Moreover, RMs infected with ORF2s mut were more susceptible to reinfection, as evidenced by the viral RNA detected in fecal samples and the expansion of HEV-specific CD8 + T cells. CONCLUSIONS: These findings indicate that ORF2s may be dispensable for viral replication in vivo but is required for long-lived antibody-mediated responses that protect against HEV re-exposure.


Subject(s)
Antibodies, Viral , Hepatitis E virus , Animals , Antibodies, Viral/metabolism , Hepatitis E virus/genetics , Macaca mulatta/metabolism , Antibody Formation , Epitopes
5.
J Virol ; 95(23): e0058521, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34523963

ABSTRACT

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.


Subject(s)
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
6.
Liver Int ; 41(12): 2866-2873, 2021 12.
Article in English | MEDLINE | ID: mdl-34392598

ABSTRACT

BACKGROUND: Hepatitis E viruses (HEV) are an underestimated global cause of enterically transmitted viral hepatitis, which may persist in immunocompromised hosts, posing a risk for progressive liver fibrosis with limited treatment options. We previously established liver-humanized mice as a model for chronic HEV infections, which can be cleared by a 2-week pegylated (peg)-Interferon(IFN)α treatment course. However, severe side effects may hamper the use of IFNα in immunocompromised transplant recipient patients. IFNλ may be a valuable alternative, as its receptor is less ubiquitously expressed. AIMS: In this study, we assess the in vitro and in vivo potency of pegIFNλ to induce innate immune signalling in liver cells and to clear a persistent HEV infection in liver-humanized mice. METHODS & RESULTS: We found that human liver cells expressed the IFNλ receptor (IFNLR1) and are responsive to pegIFNλ. Treatment with pegIFNλ of liver-humanized mice persistently infected with HEV genotype 3 showed that pegIFNλ was well tolerated. Dose escalation studies showed that although HEV was not cleared at pegIFNλ doses up to 0.12 mg/kg for a maximum of 8 weeks, a dose of 0.3 mg/kg pegIFNλ treatment resulted in complete clearance of HEV antigen and HEV RNA from the liver in 8 out of 9 liver-humanized mice. CONCLUSIONS: PegIFNλ is well tolerated in mice and leads to clearance of a persistent HEV infection in liver-humanized mice.


Subject(s)
Hepatitis E virus , Hepatitis E , Animals , Antiviral Agents/therapeutic use , Hepatitis E virus/genetics , Humans , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Mice , Receptors, Interferon/therapeutic use
7.
Cell Biosci ; 11(1): 123, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225786

ABSTRACT

BACKGROUND: The sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for hepatitis B virus (HBV). NTCP-reconstituted human hepatoma cells support HBV infection, but the infection is suboptimal and no apparent HBV spread has been observed in this system. RESULTS: We found that NTCP-reconstituted HepG2 cells were highly susceptible to HBV infection after cells were cultured in a commercial human inducible pluripotent stem cell (iPSC)-derived hepatocyte maintenance medium (HMM). The enhanced HBV infection coincided with increased NTCP expression, and was observed in six different clones of HepG2-NTCP cells. Promoter assays indicated that HMM activated the cytomegalovirus immediate-early (IE) promoter that drives the NTCP expression in the HepG2-NTCP cells. RNA-Seq analysis revealed that HMM upregulated multiple metabolic pathways. Despite highly upregulated NTCP expression by HMM, no obvious HBV spread was observed even in the presence of PEG 8000. CONCLUSIONS: Our data suggest that this particular medium could be used to enhance HBV infection in NTCP-reconstituted hepatocytes in vitro.

8.
Viruses ; 13(5)2021 05 08.
Article in English | MEDLINE | ID: mdl-34066709

ABSTRACT

Hepatitis A virus (HAV) infection is a common cause of acute viral hepatitis worldwide. Despite decades of research, the pathogenic mechanisms of hepatitis A remain incompletely understood. As the replication of HAV is noncytopathic in vitro, a widely accepted concept has been that virus-specific cytotoxic T cells are responsible for liver injury. However, accumulating evidence suggests that natural killer (NK) cells, NKT cells, and even non-HAV-specific CD8+ T cells contribute to liver damage during HAV infection. In addition, intrinsic death of virus-infected hepatocytes has been implicated as a cause of liver injury in a murine model of hepatitis A. Furthermore, genetic variations in host factors such as T cell immunoglobulin-1 (TIM1) and IL-18 binding protein (IL-18BP) have been linked to hepatitis A severity. This review summarizes the current knowledge of the mechanisms of hepatocellular injury in hepatitis A. Different mechanisms may be involved under different conditions and they are not necessarily mutually exclusive. A better understanding of these mechanisms would aid in diagnosis and treatment of diseases associated with HAV infection.


Subject(s)
Hepatitis A virus/pathogenicity , Hepatitis A/complications , Hepatocytes/pathology , Liver/injuries , Liver/virology , Animals , Carcinoma, Hepatocellular/pathology , Hepatitis A/immunology , Hepatitis A/physiopathology , Hepatitis A virus/immunology , Hepatocytes/virology , Humans , Liver/cytology , Liver Neoplasms/pathology , Mice
9.
J Hepatol ; 75(3): 557-564, 2021 09.
Article in English | MEDLINE | ID: mdl-33961939

ABSTRACT

BACKGROUND & AIMS: HEV is a significant cause of acute hepatitis globally. Some genotypes establish persistent infection when immunity is impaired. Adaptive immune mechanisms that mediate resolution of infection have not been identified. Herein, the requirement for CD8+ T cells to control HEV infection was assessed in rhesus macaques, a model of acute and persistent HEV infection in humans. METHODS: Rhesus macaques were untreated or treated with depleting anti-CD8α monoclonal antibodies before challenge with an HEV genotype (gt)3 isolate derived from a chronically infected human patient. HEV replication, alanine aminotransferase, anti-capsid antibody and HEV-specific CD4+ and CD8+ T cell responses were assessed after infection. RESULTS: HEV control in untreated macaques coincided with the onset of a neutralizing IgG response against the ORF2 capsid and liver infiltration of functional HEV-specific CD4+ and CD8+ T cells. Virus control was delayed by 1 week in CD8+ T cell-depleted macaques. Infection resolved with onset of a neutralizing IgG antibody response and a much more robust expansion of CD4+ T cells with antiviral effector function. CONCLUSIONS: Liver infiltration of functional CD8+ T cells coincident with HEV clearance in untreated rhesus macaques, and a 1-week delay in HEV clearance in CD8+ T cell-depleted rhesus macaques, support a role for this subset in timely control of virus replication. Resolution of infection in the absence of CD8+ T cells nonetheless indicates that neutralizing antibodies and/or CD4+ T cells may act autonomously to inhibit HEV replication. HEV susceptibility to multiple adaptive effector mechanisms may explain why persistence occurs only with generalized immune suppression. The findings also suggest that neutralizing antibodies and/or CD4+ T cells should be considered as a component of immunotherapy for chronic infection. LAY SUMMARY: The hepatitis E virus (HEV) is a major cause of liver disease globally. Some genetic types (genotypes) of HEV persist in the body if immunity is impaired. Our objective was to identify immune responses that promote clearance of HEV. Findings indicate that HEV may be susceptible to multiple arms of the immune response that can act independently to terminate infection. They also provide a pathway to assess immune therapies for chronic HEV infection.


Subject(s)
Hepatitis E/rehabilitation , Immunoglobulin G/pharmacology , Macaca mulatta/virology , Animals , CD8-Positive T-Lymphocytes/physiology , Disease Models, Animal , Haplorhini , Hepatitis E virus/drug effects , Hepatitis E virus/pathogenicity , Immunoglobulin G/therapeutic use , Liver/virology
10.
Future Virol ; 15(10): 707-715, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33250929

ABSTRACT

The liver is home to five known human hepatitis viruses (hepatitis A virus-hepatitis E virus). Despite being phylogenetically unrelated, these viruses replicate and spread in the liver without causing apparent cytopathic effects, and all have evolved strategies to counteract antibody-mediated inhibition of virus spread. In this review, we discuss the current understanding regarding the spread mechanisms for these viruses with an attempt to extract common principles and identify key questions for future studies.

11.
Nat Microbiol ; 5(4): 584-598, 2020 04.
Article in English | MEDLINE | ID: mdl-32015498

ABSTRACT

Internal N6-methyladenosine (m6A) modification is one of the most common and abundant modifications of RNA. However, the biological roles of viral RNA m6A remain elusive. Here, using human metapneumovirus (HMPV) as a model, we demonstrate that m6A serves as a molecular marker for innate immune discrimination of self from non-self RNAs. We show that HMPV RNAs are m6A methylated and that viral m6A methylation promotes HMPV replication and gene expression. Inactivating m6A addition sites with synonymous mutations or demethylase resulted in m6A-deficient recombinant HMPVs and virion RNAs that induced increased expression of type I interferon, which was dependent on the cytoplasmic RNA sensor RIG-I, and not on melanoma differentiation-associated protein 5 (MDA5). Mechanistically, m6A-deficient virion RNA induces higher expression of RIG-I, binds more efficiently to RIG-I and facilitates the conformational change of RIG-I, leading to enhanced interferon expression. Furthermore, m6A-deficient recombinant HMPVs triggered increased interferon in vivo and were attenuated in cotton rats but retained high immunogenicity. Collectively, our results highlight that (1) viruses acquire m6A in their RNA as a means of mimicking cellular RNA to avoid detection by innate immunity and (2) viral RNA m6A can serve as a target to attenuate HMPV for vaccine purposes.


Subject(s)
Adenosine/analogs & derivatives , DEAD Box Protein 58/genetics , Immune Evasion/genetics , Interferon-beta/genetics , Metapneumovirus/immunology , RNA, Viral/genetics , A549 Cells , Adenosine/immunology , Adenosine/metabolism , Animals , Chlorocebus aethiops , DEAD Box Protein 58/immunology , Gene Expression Regulation , Genome, Viral/immunology , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon-beta/immunology , Metapneumovirus/genetics , Metapneumovirus/growth & development , NF-kappa B/genetics , NF-kappa B/immunology , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , RNA, Viral/immunology , Receptors, Immunologic , Sigmodontinae , Signal Transduction , THP-1 Cells , Vero Cells , Virion/genetics , Virion/growth & development , Virion/immunology
12.
Adv Virus Res ; 108: 315-336, 2020.
Article in English | MEDLINE | ID: mdl-33837720

ABSTRACT

Hepatitis A virus (HAV) and hepatitis E virus (HEV) infections are the main causes for acute hepatitis worldwide. Both viruses had long been considered as nonenveloped viruses. However, recent work has uncovered that both viruses circulate in the bloodstream as membrane-cloaked, "quasi-enveloped" particles that are, surprisingly, infectious and likely the only form mediating virus spread within the host. The discovery of quasi-enveloped HAV and HEV particles has fundamentally changed the traditional view on the life cycle and pathogenesis of these viruses. However, because HAV and HEV are phylogenetically unrelated and their capsid assembly processes are quite distinct, it is not clear whether they use similar or different mechanisms for envelopment and exit. This review provides an overview of the current knowledge about the assembly and exit processes of HAV and HEV and perspectives for future studies.


Subject(s)
Hepatitis A virus/physiology , Hepatitis E virus/physiology , Viral Envelope , Virus Assembly , Virus Release , Books , Capsid , Capsid Proteins/metabolism , Cell Membrane/metabolism , Hepatitis A/blood , Hepatitis A/virology , Hepatitis A virus/genetics , Hepatitis A virus/pathogenicity , Hepatitis E/blood , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/pathogenicity , Humans
13.
Viruses ; 11(10)2019 09 20.
Article in English | MEDLINE | ID: mdl-31547135

ABSTRACT

Hepatitis E virus (HEV) infection is a major cause of acute hepatitis worldwide. It is transmitted enterically but replicates in the liver. Recent studies indicate that HEV exists in two forms: naked, nonenveloped virions that are shed into feces to mediate inter-host transmission, and membrane-cloaked, quasienveloped virions that circulate in the bloodstream to mediate virus spread within a host. Both virion types are infectious, but differ in the way they infect cells. Elucidating the entry mechanism for both virion types is essential to understand HEV biology and pathogenesis, and is relevant to the development of treatments and preventions for HEV. This review summarizes the current understanding of the cell entry mechanism for these two HEV virion types.


Subject(s)
Hepatitis E virus/physiology , Virus Internalization , Hepatitis E/metabolism , Hepatitis E/virology , Hepatitis E virus/ultrastructure , Humans , Lysosomes/metabolism , Lysosomes/virology , Receptors, Virus/metabolism , Virion/physiology , Virion/ultrastructure , Virus Uncoating
14.
Life Sci ; 221: 1-12, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30738042

ABSTRACT

Plasmin-mediated fibrinolysis at the surface of vascular endothelial cells (SVEC) plays a key role in maintaining vascular hemostasis, in which the cAMP pathway participates. After externalization to the SVEC, annexin A2 (ANXA2) serves as a platform for conversion of plasminogen to plasmin. Here we describe a regulatory role of the exchange protein directly activated by cAMP (EPAC) in ANXA2 externalization and vascular fibrinolysis. Knockout of EPAC1 in mice results in a decreased ANXA2 expression on the SVEC associated with increased fibrin deposition and fibrinolytic dysfunction. Reduced levels of EPAC1 are also found in endocardial tissues beneath atrial mural thrombi in patients. Notably, administration of recombinant ANXA2 ameliorates fibrinolytic dysfunction in the EPAC1-null mice. Mechanistically, EPAC1 regulates the SVEC plasminogen conversion depended on ANXA2. EPAC1 promotes tyrosine-23 phosphorylation of ANXA2, a prerequisite for its recruitment to the SVEC. Our data thus reveal a novel regulatory role for EPAC1 in vascular fibrinolysis.


Subject(s)
Annexin A2/metabolism , Fibrinolysis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Animals , Cell Membrane , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular , Fibrinolysin/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Homeostasis , Humans , Mice , Mice, Knockout , Phosphorylation , Plasminogen/metabolism , Proteolysis
15.
Viruses ; 11(1)2019 01 15.
Article in English | MEDLINE | ID: mdl-30650547

ABSTRACT

Hepatitis E virus (HEV) is a common cause of acute hepatitis worldwide. Current methods for evaluating the neutralizing activity of HEV-specific antibodies include immunofluorescence focus assays (IFAs) and real-time PCR, which are insensitive and operationally complicated. Here, we developed a high-throughput neutralization assay by measuring secreted pORF2 levels using an HEV antigen enzyme-linked immunosorbent assay (ELISA) kit based on the highly replicating HEV genotype (gt) 3 strain Kernow. We evaluated the neutralizing activity of HEV-specific antibodies and the sera of vaccinated individuals (n = 15) by traditional IFA and the novel assay simultaneously. A linear regression analysis shows that there is a high degree of correlation between the two assays. Furthermore, the anti-HEV IgG levels exhibited moderate correlation with the neutralizing titers of the sera of vaccinated individuals, indicating that immunization with gt 1 can protect against gt 3 Kernow infection. We then determined specificity of the novel assay and the potential threshold of neutralizing capacity using anti-HEV IgG positive sera (n = 27) and anti-HEV IgG negative sera (n = 23). The neutralizing capacity of anti-HEV IgG positive sera was significantly stronger than that of anti-HEV IgG negative. In addition, ROC curve analysis shows that the potential threshold of neutralizing capacity of sera was 8.07, and the sensitivity and specificity of the novel assay was 88.6% and 100%, respectively. Our results suggest that the neutralization assay using the antigen ELISA kit could be a useful tool for HEV clinical research.


Subject(s)
Hepatitis Antibodies/blood , Hepatitis E virus , High-Throughput Screening Assays/methods , Immunoglobulin G/blood , Neutralization Tests/methods , Viral Proteins/blood , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Hep G2 Cells , Hepatitis E/diagnosis , Hepatitis E/immunology , Humans , Immunoglobulin M/blood , Male , Middle Aged , Sensitivity and Specificity , Vaccination
16.
Article in English | MEDLINE | ID: mdl-29686040

ABSTRACT

Although hepatitis A virus (HAV) and hepatitis E virus (HEV) are both positive-strand RNA viruses that replicate in the cytoplasm of hepatocytes, there are important differences in the ways they induce and counteract host innate immune responses. HAV is remarkably stealthy because of its ability to evade and disrupt innate signaling pathways that lead to interferon production. In contrast, HEV does not block interferon production. Instead, it persists in the presence of an interferon response. These differences may provide insight into HEV persistence in immunocompromised patients, an emerging health problem in developed countries.


Subject(s)
Hepatitis A virus/immunology , Hepatitis A/immunology , Hepatitis E virus/immunology , Hepatitis E/immunology , Immunity, Innate/physiology , Animals , Cells, Cultured , Disease Models, Animal , Drug Resistance, Viral , Hepatocytes/immunology , Humans , Inflammasomes/immunology , Interferons/pharmacology , Liver/immunology
17.
Proc Natl Acad Sci U S A ; 115(18): 4773-4778, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29669922

ABSTRACT

The enterically transmitted hepatitis E virus (HEV) adopts a unique strategy to exit cells by cloaking its capsid (encoded by the viral ORF2 gene) and circulating in the blood as "quasi-enveloped" particles. However, recent evidence suggests that the majority of the ORF2 protein present in the patient serum and supernatants of HEV-infected cell culture exists in a free form and is not associated with virus particles. The origin and biological functions of this secreted form of ORF2 (ORF2S) are unknown. Here we show that production of ORF2S results from translation initiated at the previously presumed AUG start codon for the capsid protein, whereas translation of the actual capsid protein (ORF2C) is initiated at a previously unrecognized internal AUG codon (15 codons downstream of the first AUG). The addition of 15 amino acids to the N terminus of the capsid protein creates a signal sequence that drives ORF2S secretion via the secretory pathway. Unlike ORF2C, ORF2S is glycosylated and exists as a dimer. Nonetheless, ORF2S exhibits substantial antigenic overlap with the capsid, but the epitopes predicted to bind the putative cell receptor are lost. Consistent with this, ORF2S does not block HEV cell entry but inhibits antibody-mediated neutralization. These results reveal a previously unrecognized aspect in HEV biology and shed new light on the immune evasion mechanisms and pathogenesis of this virus.


Subject(s)
Epitopes/immunology , Hepatitis Antigens/immunology , Hepatitis E virus/immunology , Hepatitis E/immunology , Protein Biosynthesis/immunology , Viral Proteins/immunology , Codon, Initiator/immunology , Epitopes/genetics , Hep G2 Cells , Hepatitis Antigens/genetics , Hepatitis E/genetics , Hepatitis E/pathology , Hepatitis E virus/genetics , Humans , Protein Biosynthesis/genetics , Viral Proteins/genetics
18.
PLoS Pathog ; 13(5): e1006417, 2017 May.
Article in English | MEDLINE | ID: mdl-28558073

ABSTRACT

The RIG-I-like RNA helicase (RLR)-mediated interferon (IFN) response plays a pivotal role in the hepatic antiviral immunity. The hepatitis A virus (HAV) and the hepatitis C virus (HCV) counter this response by encoding a viral protease that cleaves the mitochondria antiviral signaling protein (MAVS), a common signaling adaptor for RLRs. However, a third hepatotropic RNA virus, the hepatitis E virus (HEV), does not appear to encode a functional protease yet persists in infected cells. We investigated HEV-induced IFN responses in human hepatoma cells and primary human hepatocytes. HEV infection resulted in persistent virus replication despite poor spread. This was companied by a type III IFN response that upregulated multiple IFN-stimulated genes (ISGs), but type I IFNs were barely detected. Blocking type III IFN production or signaling resulted in reduced ISG expression and enhanced HEV replication. Unlike HAV and HCV, HEV did not cleave MAVS; MAVS protein size, mitochondrial localization, and function remained unaltered in HEV-replicating cells. Depletion of MAVS or MDA5, and to a less extent RIG-I, also diminished IFN production and increased HEV replication. Furthermore, persistent activation of the JAK/STAT signaling rendered infected cells refractory to exogenous IFN treatment, and depletion of MAVS or the receptor for type III IFNs restored the IFN responsiveness. Collectively, these results indicate that unlike other hepatotropic RNA viruses, HEV does not target MAVS and its persistence is associated with continuous production of type III IFNs.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/immunology , Interferons/immunology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Hepatitis E/genetics , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/immunology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Interferons/genetics , Virus Replication
19.
Gastroenterology ; 152(3): 550-553.e8, 2017 02.
Article in English | MEDLINE | ID: mdl-27838287

ABSTRACT

Occult infection with hepatitis C virus (HCV) is defined as the presence of the HCV genome in either liver tissue or peripheral blood monocytes, despite constant negative results from tests for HCV RNA in serum. We investigated whether patients who maintained a sustained virologic response 12 weeks after therapy (SVR12) with direct-acting antiviral (DAA) agents for recurrent HCV infection after liver transplantation had occult HCV infections. We performed a prospective study of 134 patients with recurrent HCV infection after liver transplantation who were treated with DAAs, with or without ribavirin, from 2014 through 2016 (129 patients achieved an SVR12). In >10% of the patients who achieved SVR12 (n = 14), serum levels of aminotransferases did not normalize during or after DAA therapy, or they normalized transiently but then increased sharply after DAA therapy. Of these 14 patients, 9 were assessed for occult HCV infection by reverse transcription quantitative polymerase chain reaction. This analysis revealed that 55% of these patients (n = 5) had an occult infection, with the detection of negative strand viral genome, indicating viral replication. These findings indicate the presence of occult HCV infection in some patients with abnormal levels of serum aminotransferases, despite SVR12 to DAAs for HCV infection after liver transplantation.


Subject(s)
Alanine Transaminase/blood , Antiviral Agents/therapeutic use , Aspartate Aminotransferases/blood , Hepacivirus/genetics , Hepatitis C, Chronic/blood , Liver Transplantation , RNA, Viral/blood , Virus Replication/genetics , Benzimidazoles/therapeutic use , Carbamates , Drug Therapy, Combination , Female , Fluorenes/therapeutic use , Hepatitis C, Chronic/drug therapy , Humans , Imidazoles/therapeutic use , Limit of Detection , Male , Middle Aged , Prospective Studies , Pyrrolidines , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , Ribavirin/therapeutic use , Simeprevir/therapeutic use , Sofosbuvir/therapeutic use , Sustained Virologic Response , Valine/analogs & derivatives , Viral Load
20.
Science ; 353(6307): 1541-1545, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27633528

ABSTRACT

Hepatotropic viruses are important causes of human disease, but the intrahepatic immune response to hepatitis viruses is poorly understood because of a lack of tractable small- animal models. We describe a murine model of hepatitis A virus (HAV) infection that recapitulates critical features of type A hepatitis in humans. We demonstrate that the capacity of HAV to evade MAVS-mediated type I interferon responses defines its host species range. HAV-induced liver injury was associated with interferon-independent intrinsic hepatocellular apoptosis and hepatic inflammation that unexpectedly resulted from MAVS and IRF3/7 signaling. This murine model thus reveals a previously undefined link between innate immune responses to virus infection and acute liver injury, providing a new paradigm for viral pathogenesis in the liver.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Disease Models, Animal , Hepatitis A virus/immunology , Hepatitis A/immunology , Host-Pathogen Interactions/immunology , Liver/immunology , Mice , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Hepatitis A/pathology , Hepatitis A/virology , Hepatocytes/immunology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/immunology , Liver/pathology , Liver/virology , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction/immunology , Species Specificity , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...