Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Virol ; 96(19): e0100822, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36102649

ABSTRACT

Hepatitis E virus (HEV) infection usually results in a self-limiting acute disease; however, in infected pregnant women, it is associated with increased mortality and fulminant hepatic failure. Estrogen is known to be elevated during pregnancy, and estrogen signaling via classical estrogen receptor-ERα is known to regulate hepatocyte function and host innate immune response, including the STAT3 pathway. In this study, we investigated whether the estrogen classical signaling pathway via ERαp66 has any effect on STAT3 activation during HEV replication and HEV-induced IFN response. We first demonstrated that Huh7-S10-3 liver cells expressed the nonfunctional estrogen receptor ERαp36 isoform and lack the functional ERαp66 isoform. We further showed persistent phosphorylated-STAT3 levels in genotype 3 human HEV (Kernow P6 strain) RNA-transfected cells at later time points. In Huh7-S10-3 cells, estrogen at first-to-third trimester concentration (7.3 to 73 nM) did not significantly affect HEV replication; however, blocking of STAT3 activation led to a decrease in the HEV ORF2 protein level. Our mechanistic study revealed that STAT3 differentially regulates SOCS3 and type-III interferon (IFN) levels during HEV replication and the presence of estrogen-ERαp66 signaling stabilizes SOCS3 levels in vitro. We also demonstrate that HEV infection in pregnant and nonpregnant rabbits led to a significant increase in IFN response as measured by increased levels of IFN-stimulated-gene-15 (ISG15) mRNA levels irrespective of pregnancy status. Collectively, the results indicate that estrogen signaling and STAT3 regulate SOCS3 and IFN responses in vitro during HEV replication. The results have important implications for understanding HEV replication and HEV-induced innate immune response in pregnant women. IMPORTANCE Hepatitis E is usually a self-resolving acute disease; however, in pregnant women, HEV infection is associated with high mortality and fulminant hepatic failure. During pregnancy, estrogen levels are elevated, and in the liver, the estrogen receptor ERα is predominant and estrogen signaling is known to regulate hepatocyte metabolism and leptin-induced STAT3 levels. Viruses can module host innate immune response via STAT3. Therefore, in this study, we investigated whether STAT3 and estrogen-classical signaling via the ERαp66 pathway modulate HEV replication and HEV-induced innate immune response. We demonstrated that estrogen signaling did not affect HEV replication in human liver cells, but blocking of STAT3 activation reduced HEV capsid protein levels in human liver cells. We also showed that inhibition of STAT3 activation reduced SOCS3 levels, while the presence of the estrogen-ERαp66 signaling pathway stabilized SOCS3 levels. The results from this study will aid our understanding of the mechanism of HEV pathogenesis and immune response during pregnancy.


Subject(s)
Carcinoma, Hepatocellular , Estrogen Receptor alpha , Hepatitis E , Liver Neoplasms , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Animals , Capsid Proteins , Carcinoma, Hepatocellular/virology , Estrogen Receptor alpha/metabolism , Estrogens , Female , Hepatitis E/metabolism , Hepatitis E virus/physiology , Humans , Interferons/metabolism , Leptin/metabolism , Liver Failure, Acute/virology , Liver Neoplasms/virology , Pregnancy , RNA , RNA, Messenger , Rabbits , Receptors, Estrogen , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , Virus Replication
2.
PLoS Pathog ; 18(8): e1010798, 2022 08.
Article in English | MEDLINE | ID: mdl-36007070

ABSTRACT

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.


Subject(s)
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
3.
FASEB J ; 36(1): e22104, 2022 01.
Article in English | MEDLINE | ID: mdl-34918388

ABSTRACT

MicroRNAs (miRNAs), the non-coding RNAs of ~22 nucleotides (nt) in length, play a vital role in regulating viral replication. Hepatitis E virus (HEV), a single-stranded RNA virus, is a predominant pathogen of acute hepatitis worldwide. Virus-encoded miRNAs regulate the viral life cycle and escape from the host innate immune system. However, it is rarely known about HEV-encoded miRNA (HEV-miR-A6). In the present study, HEV-miR-A6 was screened by microarray, and further identified in vivo and in vitro. HEV-miR-A6 originated from the methylase (MeT) of HEV open reading frame 1 (ORF1) and was highly conserved in eight HEV genotypes. HEV-miR-A6 expression was growing during HEV replication, and significantly increased in acute hepatitis E patients than convalescence patients. Furthermore, HEV-miR-A6 was specifically detected in liver, spleen, kidney and colon by in situ hybridization. To identify the specificity of HEV-miR-A6, its mutants (HEV-miR-A6M1 and HEV-miR-A6M2) were constructed to change the stem-loop structure. Interestingly, over-expression of HEV-miR-A6 or HEV-miR-A6M1 significantly facilitated viral replication, while HEV-miR-A6M2, another mutant completely changed the stem-loop structure was invalid. SIRP-α, a candidate target gene of HEV-miR-A6, was activated when HEV-miR-A6 over-expressed to inhibit the phosphorylation of IRF3, and subsequently suppressed the expression of type I interferon ß (IFN-ß). The promotion of viral replication by HEV-miR-A6 further identified in vivo. Significant suppression of IFN-ß production in the serum of HEV-infected mice pre-treated with HEV-miR-A6 was observed. In summary, HEV-miR-A6 activates SIRP-α to promote viral replication by inhibition of IFN-ß expression.


Subject(s)
Gene Expression Regulation, Viral , Hepatitis E virus/physiology , Hepatitis E/metabolism , Interferon-beta/metabolism , MicroRNAs/biosynthesis , RNA, Viral/biosynthesis , Virus Replication , Female , Humans , Male , Organ Specificity
4.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502167

ABSTRACT

Hepatitis E virus (HEV) usually causes self-limiting acute hepatitis, but the disease can become chronic in immunocompromised individuals. HEV infection in pregnant women is reported to cause up to 30% mortality, especially in the third trimester. Additionally, extrahepatic manifestations like neuronal and renal diseases and pancreatitis are also reported during the course of HEV infection. The mechanism of HEV pathogenesis remains poorly understood. Innate immunity is the first line of defense triggered within minutes to hours after the first pathogenic insult. Growing evidence based on reverse genetics systems, in vitro cell culture models, and representative studies in animal models including non-human primates, has implicated the role of the host's innate immune response during HEV infection. HEV persists in presence of interferons (IFNs) plausibly by evading cellular antiviral defense. This review summarizes our current understanding of recognizing HEV-associated molecular patterns by host cell Pattern Recognition Receptors (PRRs) in eliciting innate immune response during HEV infection as well as mechanisms of virus-mediated immune evasion.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/metabolism , Hepatitis E/virology , Host-Pathogen Interactions , Receptors, Pattern Recognition/metabolism , Animals , Biomarkers , Disease Susceptibility , Gene Expression Regulation , Gene Expression Regulation, Viral , Hepatitis E/genetics , Hepatitis E/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferons/biosynthesis , Protein Binding , Receptors, Pattern Recognition/genetics
5.
Arch Virol ; 166(7): 2005-2010, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33987753

ABSTRACT

We show that ivermectin, an FDA-approved anti-parasitic drug, effectively inhibits infection with hepatitis E virus (HEV) genotypes 1 and 3 in a range of cell culture models, including hepatic and extrahepatic cells. Long-term treatment showed no clear evidence of the development of drug resistance. Gene silencing of importin-α1, a cellular target of ivermectin and a key member of the host nuclear transport complex, inhibited viral replication and largely abolished the anti-HEV effect of ivermectin.


Subject(s)
Hepatitis E virus/drug effects , Hepatitis E/drug therapy , Ivermectin/pharmacology , Virus Replication/drug effects , alpha Karyopherins/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Hepatitis E/metabolism , Hepatitis E/virology , Humans , Nuclear Proteins/metabolism
6.
Infect Genet Evol ; 92: 104882, 2021 08.
Article in English | MEDLINE | ID: mdl-33905889

ABSTRACT

With the background of association of oxidative stress and Hepatitis E virus (HEV) infection in pregnancy complications the present novel study aimed to evaluate the significance of changes in maternal homocysteine levels and the related mechanism(s) in the pathophysiology of HEV related pregnancy complications and negative outcomes. Term delivery (TD, N = 194) and HEV-IgM positive pregnancy cases [N = 109] were enrolled. Serum and placental homocysteine levels were evaluated by ELISA and immunofluorescence and in turn correlated with serum Vitamin B12 levels. Distribution of variant MTHFR C➔T and TYMS1494del6bp genotyping were studied by PCR-RFLP. Differential folate receptor alpha (FR-α) expression in placenta was evaluated by real-time PCR and immunofluorescence respectively. The HEV viral load was significantly higher in both FHF and AVH cases. Higher serum homocysteine levels was associated with preterm delivery (PTD) and fetal death in HEV infected cases and was significantly inversely correlated with serum VitaminB12 levels in HEV cases. Placental homocysteine expression was upregulated in HEV cases, and in cases with negative pregnancy outcome. A Homocysteine level was associated with MTHFR C677T status. Genetic alterations in folate pathway was associated with increased risk of PTD in HEV infected pregnancy cases, disease severity, and negative pregnancy outcome in AVH and FHF groups. FR-α expression was downregulated in placental tissues of HEV infected pregnancy.Placental stress caused by HEV inflicted increased homocysteine due to alterations in maternal vitamin B12 levels and folate pathway components is detrimental mechanism in PTD and negative pregnancy outcome in HEV infected pregnancy cases and holds prognostic and therapeutic significance.


Subject(s)
Hepatitis E/metabolism , Hepevirus/physiology , Homocysteine/metabolism , Oxidative Stress , Pregnancy Complications, Infectious/metabolism , Adult , Female , Hepatitis E/virology , Humans , India , Pregnancy , Pregnancy Complications, Infectious/virology , Young Adult
8.
J Immunol Res ; 2020: 6027361, 2020.
Article in English | MEDLINE | ID: mdl-33294465

ABSTRACT

BACKGROUND AND AIMS: The involvement of cellular immunity in the development of hepatitis E virus (HEV) infection is rare. We aimed to study the roles of viral load and Th cell responses in acute hepatitis E (AHE) and HEV-related acute liver failure (HEV-ALF). METHODS: We evaluated viral load and Th1/Th2 cytokine levels in 34 patients with HEV infection, including 17 each with AHE or HEV-ALF. Seventeen healthy controls (HCs) were also included who were negative for anti-HEV IgM and IgG. RESULTS: There was no significant difference in viral load and HEV RNA in the AHE and HEV-ALF groups (both P > 0.05). The Th lymphocyte levels (CD3+, CD4+) in the AHE and HEV-ALF groups were significantly higher than those in the HC group (both P < 0.05), but there was no significant difference between the AHE and HEV-ALF groups (P > 0.05). Both IFN-γ and IL-10 showed gradual upward trend from the HC group to the AHE (both P < 0.01), but IFN-γ showed a sharp downward trend from the AHE group to the HEV-ALF group (P < 0.01) and IL-4 showed gradual upward trend from the AHE group to the HEV-ALF group (P < 0.01).There was no significant difference in Th1 and Th2 cytokines between the HEV RNA(+) group and HEV RNA(-) group (all P > 0.05). Th2 bias was observed from the AHE (ratio = 58.65) to HEV-ALF (ratio = 1.20) groups. The level of IFN-γ was associated with the outcome of HEV-ALF patients. CONCLUSIONS: HEV viral load was not associated with aggravation of AHE, and the HEV-ALF patients showed significant Th2 bias, which may be involved in the aggravation of AHE.


Subject(s)
Cytokines/metabolism , Hepatitis E/immunology , Hepatitis E/metabolism , Liver Failure, Acute/etiology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Antibody Specificity/immunology , Biomarkers , Disease Susceptibility , Female , Hepatitis Antibodies/immunology , Hepatitis E/complications , Hepatitis E/virology , Hepatitis E virus/immunology , Humans , Liver Failure, Acute/diagnosis , Male , RNA, Viral , Severity of Illness Index , Th1 Cells/pathology , Th2 Cells/pathology , Viral Load
9.
Emerg Microbes Infect ; 9(1): 2697-2705, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33251979

ABSTRACT

The pathogenicity of each hepatitis E virus (HEV) genotypes/subtypes may be different. This study aimed to investigate the infectivity and pathogenicity of different HEV genotypes/subtypes from different mammalian sources especially human in rabbits, and to assess whether rabbits are an appropriate animal model to study different HEV genotypes/subtypes. Thirty-seven rabbits were randomly divided into nine groups and inoculated with eight different HEV strains, including human-derived HEV3b (hHEV-3b), hHEV-4a, hHEV-4d and hHEV-4h, swine-derived HEV4d (sHEV-4d) and sHEV-4h, rabbit-derived HEV3 (HEV-3ra) and camel-derived HEV8. HEV RNA, antigen, anti-HEV and alanine aminotransferase (ALT) in serum or/and feces were monitored weekly. One rabbit from each group was euthanized at seven weeks post inoculation and the liver specimens were taken for histopathological analysis and immunofluorescence staining of HEV ORF2 proteins. hHEV-4d, sHEV-4d and HEV-3ra infections were successfully established in rabbits and typical acute hepatitis symptoms were observed, including viraemia/antigenemia, fecal virus/antigen shedding, elevated ALT level and liver histopathological changes. One rabbit infected with HEV-3ra showed chronic infection. hHEV-4d and sHEV-4d are less infectious and pathogenic than HEV-3ra in rabbits. hHEV-3b and HEV8 only caused inapparent infection in rabbits as 60% (3/5) and 20% (1/5) of the rabbits seroconverted to anti-HEV, respectively. No obvious signs of HEV infection in rabbits inoculated with hHEV-4a, hHEV-4h and sHEV-4h. The infectivity and pathogenicity of different HEV genotypes/subtypes in rabbits is different, which may be related to the species specificity of HEV. Rabbit can be used as an animal model for the study of HEV-3ra and more importantly human HEV-4d.


Subject(s)
Alanine Transaminase/metabolism , Hepatitis Antibodies/metabolism , Hepatitis E virus/pathogenicity , Hepatitis E/metabolism , Liver/metabolism , RNA, Viral/genetics , Viral Proteins/metabolism , Alanine Transaminase/blood , Animals , Camelus , Disease Models, Animal , Genotype , Hepatitis Antibodies/blood , Hepatitis E/blood , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Humans , Liver/virology , Phylogeny , Rabbits , Random Allocation , Species Specificity , Swine , Virulence
10.
Viruses ; 12(8)2020 07 30.
Article in English | MEDLINE | ID: mdl-32751663

ABSTRACT

Exosomes seem to play an important role in hepatits C virus (HCV) and hepatitis E virus (HEV) infection by shielding their cargo from the host immune responses, with microRNAs being key exosomal components. Little is known about their involvement in a mixed HCV/HEV infection or at the early stages of infection, such as in asymptomatic blood donors (BDs). To obtain preliminary data, we have compared the exosomal microRNA expression profiles in four each of HCV RNA-positive, HEV RNA-positive and negative blood donors and four patients, one of whom was a rare patient with HCV/HEV co-infection. Exosomes were purified from sera by a combination of a precipitation and density gradient centrifugation and exosomal microRNA was analysed using Taqman array cards. Out of 33 deregulated miRNAs, miR-885-5p and miR-365 were upregulated in HCV BDs, miR-627-5p was downregulated in HCV BD and miR-221 was downregulated in HCV patients and BDs. In HEV infection, miR-526b appeared specifically downregulated. Six miRNAs (miR-628-3p, miR-194, miR-151-3p, miR-512-3p, miR-335 and miR-590) indicated a potential involvement in both infections. First time preliminary data on pre- and post-antiviral treatment exosomal microRNA profiles of the HEV/HCV co-infected patient revealed a pool of 77 upregulated and 43 downregulated miRNAs to be further investigated for their potential roles in these viral infections.


Subject(s)
Coinfection , Exosomes/metabolism , Hepatitis C/genetics , Hepatitis E/genetics , MicroRNAs/genetics , Aged , Antiviral Agents/therapeutic use , Blood Donors , Exosomes/genetics , Female , Gene Expression Profiling , Hepatitis C/complications , Hepatitis C/drug therapy , Hepatitis C/metabolism , Hepatitis E/complications , Hepatitis E/drug therapy , Hepatitis E/metabolism , Humans , Male , MicroRNAs/metabolism , Middle Aged , Pilot Projects
11.
Clin Microbiol Infect ; 26(7): 828-832, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32251845

ABSTRACT

BACKGROUND: Hepatitis E virus (HEV) is a common cause of viral hepatitis worldwide. Previously considered a disease of the developing world, it is increasingly recognized that locally acquired HEV infection is common in industrialized countries. OBJECTIVES: The aim was to highlight the changing epidemiology of HEV infection, particularly in the developed world, and inform clinicians of the diverse clinical presentations and extra-hepatic complications associated with the virus. SOURCES: References for this review were identified through searches of MEDLINE/PubMed, and Google Scholar, up to January 2020. Searches were restricted to articles published in English. CONTENT: Hepatitis E virus is an under-recognized, emerging pathogen with important implications for public health in both the developing and developed world. The number of cases reported in resource-rich settings is increasing, in part due to improved case ascertainment but also as a result of increased incidence in some countries. The reasons behind these epidemiological shifts are not currently known. Chronic HEV infection has been reported in immunocompromised patients. A range of extra-hepatic manifestations have also been reported, most notably neurological and renal complications. There is evidence to suggest a causal link with Guillain-Barré syndrome, neuralgic amyotrophy and encephalitis/myelitis. Glomerular disease has been reported in the context of both acute and chronic infection. IMPLICATIONS: HEV should be included in non-invasive liver screens and considered in the differentials for patients presenting with alanine aminotransferase elevation, suspected drug-induced liver injury or decompensated liver disease. Any patients with acute neurological injury and deranged liver function should be tested for hepatitis E, and all patients presenting with Guillain-Barré syndrome or neuralgic amyotrophy should be tested regardless of liver enzymes. Immunocompromised patients with persistently raised liver enzymes should be tested with molecular techniques and offered annual routine screening.


Subject(s)
Alanine Transaminase/metabolism , Hepatitis E/diagnosis , Hepatitis E/epidemiology , Developed Countries , Diagnosis, Differential , Early Diagnosis , Global Health , Hepatitis E/metabolism , Humans , Immunocompromised Host , Incidence
12.
Proc Natl Acad Sci U S A ; 117(3): 1731-1741, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31896581

ABSTRACT

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and the leading cause for acute viral hepatitis worldwide. The virus is classified as a member of the genus Orthohepevirus A within the Hepeviridae family. Due to the absence of a robust cell culture model for HEV infection, the analysis of the viral life cycle, the development of effective antivirals and a vaccine is severely limited. In this study, we established a protocol based on the HEV genotype 3 p6 (Kernow C-1) and the human hepatoma cell lines HepG2 and HepG2/C3A with different media conditions to produce intracellular HEV cell culture-derived particles (HEVcc) with viral titers between 105 and 106 FFU/mL. Viral titers could be further enhanced by an HEV variant harboring a mutation in the RNA-dependent RNA polymerase. These HEVcc particles were characterized in density gradients and allowed the trans-complementation of subgenomic reporter HEV replicons. In addition, in vitro produced intracellular-derived particles were infectious in liver-humanized mice with high RNA copy numbers detectable in serum and feces. Efficient infection of primary human and swine hepatocytes using the developed protocol could be observed and was inhibited by ribavirin. Finally, RNA sequencing studies of HEV-infected primary human hepatocytes demonstrated a temporally structured transcriptional defense response. In conclusion, this robust cell culture model of HEV infection provides a powerful tool for studying viral-host interactions that should facilitate the discovery of antiviral drugs for this important zoonotic pathogen.


Subject(s)
Hepatitis E virus/genetics , Hepatitis E virus/physiology , Hepatitis E/metabolism , Hepatocytes/virology , Animals , Antiviral Agents/pharmacology , Carcinoma, Hepatocellular , Cell Culture Techniques , Cell Line, Tumor , Genotype , Hep G2 Cells , Hepatitis E/virology , Hepatitis E virus/drug effects , Humans , Liver Neoplasms/drug therapy , Mice , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Replicon , Ribavirin/metabolism , Swine , Viral Load , Virus Replication
13.
IUBMB Life ; 72(4): 624-631, 2020 04.
Article in English | MEDLINE | ID: mdl-31769934

ABSTRACT

Viral replication and related protein expression inside the host cells, and host antiviral immune responses can lead to the occurrence of diverse diseases. With the outbreak of viral infection, a large number of newly diagnosed and died patients infected with various viruses are still reported every year. Viral infection has already been one of the major global public health issues and lead to huge economic and social burdens. Studying of viral pathogenesis is a very important way to find methods for prevention, diagnosis, and cure of viral infection; more evidence has confirmed that major vault protein (MVP) is closely associated with viral infection and pathogenesis, and this review is intended to provide a broad relationship between viruses and MVP to stimulate the interest of related researchers.


Subject(s)
Host-Pathogen Interactions/physiology , Vault Ribonucleoprotein Particles/physiology , Virus Diseases/virology , Antiviral Agents/pharmacology , Cystatin B/metabolism , HIV Infections/metabolism , HIV Infections/virology , Hepatitis E/drug therapy , Hepatitis E/metabolism , Humans , Inflammation/metabolism , Inflammation/virology , Interferon Type I/metabolism , Triterpenes/pharmacology , Virus Replication
14.
Clin Mol Hepatol ; 26(1): 16-23, 2020 01.
Article in English | MEDLINE | ID: mdl-31601068

ABSTRACT

Hepatitis E virus (HEV) is a significant health problem with approximately 20 million individuals infected annually. HEV infection has been associated with a wide spectrum of extrahepatic manifestations, including neurological, hematological and renal disorders. Guillain-Barré syndrome and neuralgic amyotrophy are the most frequent neurological manifestations. In addition, HEV infection has been observed with other neurological diseases, such as encephalitis, myelitis and Bell's palsy. Hematologic manifestations include anemia due to glucose-6-phospate dehydrogonase deficiency, autoimmune hemolytic anemia and severe thrombocytopenia. Membranoproliferative glomerulonephritis and relapse IgA nephropathy with or without coexisting cryoglobulinemia appear to be the most common renal injuries related with HEV infection. Also, HEV infection has been associated with acute pancreatitis and other immune-mediated manifestations, such as arthritis and myocarditis. However, the pathophysiologic mechanisms of HEV-related extrahepatic manifestations are still largely unclear.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/pathology , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/pathology , Hematologic Diseases/etiology , Hematologic Diseases/pathology , Hepatitis E/complications , Hepatitis E/metabolism , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Nervous System Diseases/etiology , Nervous System Diseases/pathology , Pancreatitis/etiology , Pancreatitis/pathology
15.
Viruses ; 11(11)2019 11 11.
Article in English | MEDLINE | ID: mdl-31717991

ABSTRACT

Hepatitis E virus (HEV) causes predominantly acute and self-limiting hepatitis. However, in HEV-infected pregnant women, the case fatality rate because of fulminant hepatitis can be up to 30%. HEV infection is zoonotic for some genotypes. The HEV genome contains three open reading frames: ORF1 encodes the non-structural polyprotein involved in viral RNA replication; ORF2 encodes the capsid protein; ORF3 encodes a small multifunctional protein. Interferons (IFNs) play a significant role in the early stage of the host antiviral response. In this study, we discovered that the capsid protein antagonizes IFN induction. Mechanistically, the capsid protein blocked the phosphorylation of IFN regulatory factor 3 (IRF3) via interaction with the multiprotein complex consisting of mitochondrial antiviral-signaling protein (MAVS), TANK-binding kinase 1 (TBK1), and IRF3. The N-terminal domain of the capsid protein was found to be responsible for the inhibition of IRF3 activation. Further study showed that the arginine-rich-motif in the N-terminal domain is indispensable for the inhibition as mutations of any of the arginine residues abolished the blockage of IRF3 phosphorylation. These results provide further insight into HEV interference with the host innate immunity.


Subject(s)
Capsid Proteins/metabolism , Hepatitis E virus/physiology , Hepatitis E/metabolism , Hepatitis E/virology , Host-Pathogen Interactions , Interferons/biosynthesis , Protein Interaction Domains and Motifs , Capsid Proteins/genetics , Genotype , Humans , Interferon Regulatory Factor-3/metabolism , Interferons/chemistry , Models, Biological , Nerve Growth Factors , Phosphorylation , Poly I-C/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism
16.
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
17.
J Microbiol Biotechnol ; 29(7): 1137-1143, 2019 Jul 28.
Article in English | MEDLINE | ID: mdl-31216792

ABSTRACT

Hepatitis E virus (HEV) accounts for 20 million infections in humans worldwide. In most cases, the infections are self-limiting while HEV genotype 1 infection cases may lead to lethal infections in pregnant women (~ 20% fatality). The lack of small animal models has hampered detailed analysis of virus-host interactions and HEV-induced pathology. Here, by employing a recently developed culture-adapted HEV, we demonstrated that methyltransferase, a nonstructural protein, strongly inhibits melanoma differentiation-associated gene 5 (MDA5)- mediated activation of type I interferon responses. Compared to uninfected controls, HEVinfected cells display significantly lower levels of IFN-ß promoter activation when assessed by luciferase assay and RT-PCR. HEV genome-wide screening showed that HEV-encoded methyltransferase (MeT) strongly inhibits MDA5-mediated transcriptional activation of IFN-ß and NF-κB in a dose-responsive manner whether or not it is expressed in the presence/ absence of a tag fused to it. Taken together, current studies clearly demonstrated that HEV MeT is a novel antagonist of MDA5-mediated induction of IFN-ß signaling.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/metabolism , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Methyltransferases/metabolism , Viral Proteins/metabolism , A549 Cells , HEK293 Cells , Host-Pathogen Interactions , Humans , Interferon Type I/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Transcription, Genetic
18.
J Gen Virol ; 100(3): 471-483, 2019 03.
Article in English | MEDLINE | ID: mdl-30702423

ABSTRACT

Hepatitis E virus (HEV) induces interferons and regulates the induction of interferon-stimulated genes (ISGs) in the host cell. HEV infection has been shown to promote the expression of different ISGs, such as ISG15, IFIT1, MX1, RSAD2/Viperin and CxCL10, in cell culture and animal models. Interferon-induced protein with tetratricopeptide repeat 1 (IFIT1) is an ISG-encoded protein that inhibits the translation of viral RNA, having 5'-triphosphate or the mRNA lacking 2'-O-methylation on the 5'cap. In this study, we found that IFIT1 binds to HEV RNA to inhibit its translation. HEV replication is also restricted in hepatoma cells with overexpressed IFIT1. However, despite this binding of IFIT1 to HEV RNA, HEV successfully replicates in hepatoma cells in the infection scenario. In an effort to identify the underlying mechanism, we found that HEV RNA-dependent RNA polymerase (RdRp) binds to IFIT1, thereby protecting the viral RNA from IFIT1-mediated translation inhibition. RdRp sequesters IFIT1, resulting in the successful progression of viral replication in the infected cells. Thus, we discovered a distinct pro-viral role of HEV RdRp that is crucial for successful infection in the host, and propose a unique mechanism developed by HEV to overcome IFIT1-mediated host immune response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Viral , Hepatitis E virus/enzymology , Hepatitis E/metabolism , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Hepatitis E/genetics , Hepatitis E/virology , Hepatitis E virus/genetics , Host-Pathogen Interactions , Humans , Protein Biosynthesis , RNA, Viral/genetics , RNA-Binding Proteins/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics
19.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29321328

ABSTRACT

Hepatitis E virus (HEV) is a clinically important positive-sense RNA virus. The ORF1 of HEV encodes a nonstructural polyprotein of 1,693 amino acids. It is not clear whether the ORF1 polyprotein (pORF1) is processed into distinct enzymatic domains. Many researchers have attempted to understand the mechanisms of pORF1 processing. However, these studies gave various results and could never convincingly establish the mechanism of pORF1 processing. In this study, we demonstrated the possible role of thrombin and factor Xa in pORF1 processing. We observed that the HEV pORF1 polyprotein bears conserved cleavage sites of thrombin and factor Xa. Using a reverse genetics approach, we demonstrated that an HEV replicon having mutations in the cleavage sites of either thrombin or factor Xa could not replicate efficiently in cell culture. Further, we demonstrated in vitro processing when we incubated recombinant pORF1 fragments with thrombin, and we observed the processing of pORF1 polyprotein. The treatment of a liver cell line with a serine protease inhibitor as well as small interfering RNA (siRNA) knockdown of thrombin and factor Xa resulted in significant reduction in the replication of HEV. Thrombin and factor Xa have been well studied for their roles in blood clotting. Both of these proteins are believed to be present in the active form in the blood plasma. Interestingly, in this report, we demonstrated the presence of biologically active thrombin and factor Xa in a liver cell line. The results suggest that factor Xa and thrombin are essential for the replication of HEV and may be involved in pORF1 polyprotein processing of HEV.IMPORTANCE Hepatitis E virus (HEV) causes a liver disorder called hepatitis in humans, which is mostly an acute and self-limiting infection in adults. A high mortality rate of about 30% is observed in HEV-infected pregnant women in developing countries. There is no convincing opinion about HEV ORF1 polyprotein processing owing to the variability of study results obtained so far. HEV pORF1 has cleavage sites for two host cellular serine proteases, thrombin and factor Xa, that are conserved among HEV genotypes. For the first time, this study demonstrated that thrombin and factor Xa cleavage sites on HEV pORF1 are obligatory for HEV replication. Intracellular biochemical activities of the said serine proteases are also essential for efficient HEV replication in cell culture and must be involved in pORF1 processing. This study sheds light on the presence and roles of clotting factors with respect to virus replication in the cells.


Subject(s)
Factor Xa/metabolism , Hepatitis E virus/physiology , Hepatitis E/metabolism , Polyproteins/metabolism , Proteolysis , Thrombin/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Cell Line, Tumor , Hepatitis E/genetics , Hepatitis E/pathology , Humans
20.
Gastroenterology ; 154(1): 211-223.e8, 2018 01.
Article in English | MEDLINE | ID: mdl-28958858

ABSTRACT

BACKGROUND & AIMS: Hepatitis E virus (HEV) infection is a major cause of acute hepatitis worldwide. Approximately 2 billion people live in areas endemic for HEV and are at risk of infection. The HEV genome encodes 3 proteins, including the ORF2 capsid protein. Detailed analyses of the HEV life cycle has been hampered by the lack of an efficient viral culture system. METHODS: We performed studies with gt3 HEV cell culture-produced particles and patient blood and stool samples. Samples were fractionated on iodixanol gradients and cushions. Infectivity assays were performed in vitro and in human liver chimeric mice. Proteins were analyzed by biochemical and proteomic approaches. Infectious particles were analyzed by transmission electron microscopy. HEV antigen levels were measured with the Wantaï enzyme-linked immunosorbent assay. RESULTS: We developed an efficient cell culture system and isolated HEV particles that were infectious in vitro and in vivo. Using transmission electron microscopy, we defined the ultrastructure of HEV cell culture-produced particles and particles from patient sera and stool samples. We also identified the precise sequence of the infectious particle-associated ORF2 capsid protein. In cultured cells and in samples from patients, HEV produced 3 forms of the ORF2 capsid protein: infectious/intracellular ORF2 (ORF2i), glycosylated ORF2 (ORF2g), and cleaved ORF2 (ORF2c). The ORF2i protein associated with infectious particles, whereas the ORF2g and ORF2c proteins were massively secreted glycoproteins not associated with infectious particles. ORF2g and ORF2c were the most abundant antigens detected in sera from patients. CONCLUSIONS: We developed a cell culture system and characterized HEV particles; we identified 3 ORF2 capsid proteins (ORF2i, ORF2g, and ORFc). These findings will advance our understanding of the HEV life cycle and improve diagnosis.


Subject(s)
Capsid Proteins/isolation & purification , Hepatitis E virus/physiology , Hepatitis E/metabolism , Viral Proteins/isolation & purification , Animals , Cell Culture Techniques , Disease Models, Animal , Hepatitis E/etiology , Hepatitis E/pathology , Hepatocytes , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...