Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Cell Mol Life Sci ; 80(11): 326, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37833515

RESUMO

The hepatitis E virus (HEV) is an underestimated RNA virus of which the viral life cycle and pathogenicity remain partially understood and for which specific antivirals are lacking. The virus exists in two forms: nonenveloped HEV that is shed in feces and transmits between hosts; and membrane-associated, quasi-enveloped HEV that circulates in the blood. It is suggested that both forms employ different mechanisms for cellular entry and internalization but little is known about the exact mechanisms. Interestingly, the membrane of enveloped HEV is enriched with phosphatidylserine, a natural ligand for the T-cell immunoglobulin and mucin domain-containing protein 1 (TIM1) during apoptosis and involved in 'apoptotic mimicry', a process by which viruses hijack the apoptosis pathway to promote infection. We here investigated the role of TIM1 in the entry process of HEV. We determined that HEV infection with particles derived from culture supernatant, which are cloaked by host-derived membranes (eHEV), was significantly impaired after knockout of TIM1, whereas infection with intracellular HEV particles (iHEV) was unaffected. eHEV infection was restored upon TIM1 expression; and enhanced after ectopic TIM1 expression. The significance of TIM1 during entry was further confirmed by viral binding assay, and point mutations of the PS-binding pocket diminished eHEV infection. In addition, Annexin V, a PS-binding molecule also significantly reduced infection. Taken together, our findings support a role for TIM1 in eHEV-mediated cell entry, facilitated by the PS present on the viral membrane, a strategy HEV may use to promote viral spread throughout the infected body.


Assuntos
Vírus da Hepatite E , Vírus , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Internalização do Vírus , Receptores de Superfície Celular/metabolismo
2.
Molecules ; 28(6)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36985647

RESUMO

Hepatitis E virus (HEV) is the notable causative agent of acute and chronic hepatic, renal, pancreatic, neurological, and hematopoietic blood cell infections with high risk in immunocompromised patients. Hepatic failure is mostly documented among adults, pregnant women, and patients with preexisting liver disease. HEV is a positive sense RNA virus of 7.2 kb genome size with typically three open reading frames (ORFs) which play essential roles in viral replication, genome assembly, and transcription. The mutational substitution in the viral RNA genome makes more it difficult to understand the actual relationship in the host-virus association. ORFs of HEV encode different structural and non-structural proteins and one of them is the capsid protein which is coded by ORF2. The capsid protein mediates the encapsulation of the viral genome as well as being involved in virion assembly. In the current study, the ligand-based docking approach was employed to inhibit the active amino acids of the viral capsid protein. Depending upon S-score, ADMET profiling, and drug scanning, the top ten tetrapeptides were selected as potential drug candidates with no toxicity counter to HEV receptor protein. The S-score or docking score is a mathematical function which predicts the binding affinities of docked complexes. The binding affinity of the predicted drug-target complexes helps in the selectivity of the desired compound as a potential drug. The best two selected peptides (i.e., TDGH with S-score of -8.5 and EGDE with S-score of -8.0) interacted with the active site amino acids of the capsid protein (i.e., Arg399, Gln420, and Asp444). The molecular dynamics simulations of RMSD trajectories of TDGH-capsid protein and EDGE-capsid protein have revealed that both docked complexes were structurally stable. The study revealed that these tetrapeptides would serve as strong potential inhibitors and a starting point for the development of new drug molecules against the HEV capsid protein. In future, in vivo studies are needed to explore selected peptides as potential drug candidates.


Assuntos
Vírus da Hepatite E , Gravidez , Humanos , Feminino , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Proteínas do Capsídeo/metabolismo , Peptídeos/metabolismo , Fígado/metabolismo , Aminoácidos/metabolismo
3.
Biomol NMR Assign ; 17(1): 1-8, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36272047

RESUMO

The genome of Hepatitis E virus (HEV) is 7.2 kilobases long and has three open reading frames. The largest one is ORF1, encoding a non-structural protein involved in the replication process, and whose processing is ill-defined. The ORF1 protein is a multi-modular protein which includes a macro domain (MD). MDs are evolutionarily conserved structures throughout all kingdoms of life. MDs participate in the recognition and removal of ADP-ribosylation, and specifically viral MDs have been identified as erasers of ADP-ribose moieties interpreting them as important players at escaping the early stages of host-immune response. A detailed structural analysis of the apo and bound to ADP-ribose state of the native HEV MD would provide the structural information to understand how HEV MD is implicated in virus-host interplay and how it interacts with its intracellular partner during viral replication. In the present study we present the high yield expression of the native macro domain of HEV and its analysis by solution NMR spectroscopy. The HEV MD is folded in solution and we present a nearly complete backbone and sidechains assignment for apo and bound states. In addition, a secondary structure prediction by TALOS + analysis was performed. The results indicated that HEV MD has a α/ß/α topology very similar to that of most viral macro domains.


Assuntos
Adenosina Difosfato Ribose , Vírus da Hepatite E , Adenosina Difosfato Ribose/metabolismo , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Ressonância Magnética Nuclear Biomolecular , Espectroscopia de Ressonância Magnética
4.
Virology ; 578: 128-140, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36527931

RESUMO

Hepatitis E virus (HEV), a major cause of acute viral hepatitis, is a single-stranded, positive-sense RNA virus. As such, it encodes a 1700-residue replication polyprotein pORF1 that directs synthesis of new viral RNA in infected cells. Here we report extensive modeling with AlphaFold2 of the full-length pORF1, and its production by in vitro translation. From this, we give a detailed update on the breakdown into domains of HEV pORF1. We also provide evidence that pORF1's N-terminal domain is likely to oligomerize to form a dodecameric pore, homologously to what has been described for Chikungunya virus. Beyond providing accurate folds for its five domains, our work highlights that there is no canonical protease encoded in pORF1 and that flexibility in several functionally important regions rather than proteolytic processing may serve to regulate HEV RNA synthesis.


Assuntos
Vírus da Hepatite E , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Poliproteínas/genética , Poliproteínas/metabolismo , Proteólise , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Replicação Viral/fisiologia , RNA Viral/genética , RNA Viral/metabolismo
5.
Genes (Basel) ; 14(1)2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36672782

RESUMO

The hepatitis E virus (HEV) is a long-ignored virus that has spread globally with time. It ranked 6th among the top risk-ranking viruses with high zoonotic spillover potential; thus, considering its viral threats is a pressing priority. The molecular pathophysiology of HEV infection or the underlying cause is limited. Therefore, we incorporated an unbiased, systematic methodology to get insights into the biological heterogeneity associated with the HEV. Our study fetched 93 and 2016 differentially expressed genes (DEGs) from chronic HEV (CHEV) infection in kidney-transplant patients, followed by hub module selection from a weighted gene co-expression network (WGCN). Most of the hub genes identified in this study were associated with interferon (IFN) signaling pathways. Amongst the genes induced by IFNs, the 2'-5'-oligoadenylate synthase 3 (OAS3) protein was upregulated. Protein-protein interaction (PPI) modular, functional enrichment, and feed-forward loop (FFL) analyses led to the identification of two key miRNAs, i.e., miR-222-3p and miR-125b-5p, which showed a strong association with the OAS3 gene and TRAF-type zinc finger domain containing 1 (TRAFD1) transcription factor (TF) based on essential centrality measures. Further experimental studies are required to substantiate the significance of these FFL-associated genes and miRNAs with their respective functions in CHEV. To our knowledge, it is the first time that miR-222-3p has been described as a reference miRNA for use in CHEV sample analyses. In conclusion, our study has enlightened a few budding targets of HEV, which might help us understand the cellular and molecular pathways dysregulated in HEV through various factors. Thus, providing a novel insight into its pathophysiology and progression dynamics.


Assuntos
Vírus da Hepatite E , MicroRNAs , Humanos , 2',5'-Oligoadenilato Sintetase/genética , Nucleotídeos de Adenina , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Multiômica
6.
J Hepatol ; 76(1): 46-52, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34461207

RESUMO

BACKGROUND AND AIMS: Immunocompromised patients are at risk of chronic hepatitis E which can be acquired by blood transfusions. Currently, screening of blood donors (BDs) for HEV RNA with a limit of detection (LOD) of 2,000 IU/ml is required in Germany. However, this may result in up to 440,000 IU of HEV RNA in blood products depending on their plasma volume. We studied the residual risk of transfusion-transmitted (tt) HEV infection when an LOD of 2,000 IU/ml is applied. METHODS: Highly sensitive individual donor testing for HEV RNA on the Grifols Procleix Panther system (LOD 7.89 IU/ml) was performed. HEV loads were quantified by real-time PCR. RESULTS: Of 16,236 donors, 31 (0.19%) were HEV RNA positive. Three BDs had viral loads between 710 and 2,000 IU/ml, which pose a significant risk of tt hepatitis E with any type of blood product. Eight BDs had viral loads of >32 to 710 IU/ml, which pose a risk of tt hepatitis E with platelet or plasma transfusions because of their higher plasma volume compared to red blood cell concentrates. Eight of these 11 potentially infectious BDs were seronegative for HEV, indicating a recent infection. Only 8 of 31 donors had viral loads >2,000 IU/ml that would also have been detected by the required screening procedure and 12 had very low HEV loads (<32 IU/ml). CONCLUSIONS: Screening of BDs with an LOD of 2,000 IU/ml reduced the risk of tt HEV infection by about 73% for red blood cell concentrates but by just 42% for platelet and fresh frozen plasma transfusions. Single donor screening (LOD <32 IU/ml) should lead to an almost 100% risk reduction. LAY SUMMARY: Immunocompromised patients, such as solid organ or hematopoietic stem cell recipients, are at risk of chronic hepatitis E, which can be acquired via blood transfusions. The risk of transfusion-transmitted hepatitis E in these patients may not be sufficiently controlled by (mini-)pool hepatitis E virus RNA screening of blood donors. Single donor screening should be considered to improve the safety of blood products.


Assuntos
Transfusão de Sangue/normas , Hepatite E/transmissão , Reação Transfusional/diagnóstico , Adulto , Transfusão de Sangue/métodos , Transfusão de Sangue/estatística & dados numéricos , Seleção do Doador/normas , Seleção do Doador/estatística & dados numéricos , Feminino , Alemanha , Hepatite E/sangue , Vírus da Hepatite E/metabolismo , Vírus da Hepatite E/patogenicidade , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Medição de Risco/métodos , Medição de Risco/estatística & dados numéricos , Estatísticas não Paramétricas , Reação Transfusional/fisiopatologia
7.
PLoS One ; 16(7): e0255054, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34293039

RESUMO

BACKGROUND: Hepatitis E virus (HEV) infection is endemic in Bangladesh and there are occasional outbreaks. The molecular characteristics and pathogenesis of endemic and outbreak HEV strains are poorly understood. We compared the genetic relatedness and virulence associated mutations of endemic HEV strains with outbreak strains. METHODS: We analyzed systematically collected serum samples from HEV immunoglobulin M (IgM) positive patients attended at Bangabandhu Sheikh Mujib Medical University, Dhaka from August 2013 to June 2015. HEV RNA positive samples were subjected to whole genome sequencing. Genotype and subtype of the strains were determined by phylogenetic analysis. Virulence associated mutations e.g. acute viral hepatitis (AVH), fulminant hepatic failure (FHF), chronic hepatitis, ribavirin treatment failure (RTF), B and T cell neutralization epitopes were determined. RESULTS: 92 HEV immunoglobulin M (IgM) antibody positive plasma samples (43 in 2013-2014 and 49 in 2014-2015) were studied. 77.1% (70/92) of the samples were HEV RNA positive. A 279 bp open reading frame (ORF) 2 and ORF 3 sequence was obtained from 54.2% (38/70) of the strains. Of these 38 strains, whole genome sequence (WGS) was obtained from 21 strains. In phylogenetic analysis of 38 (279 bp) sequence all HEV sequences belonged to genotype 1 and subtype 1a. Further phylogenetic analysis of 21 HEV WGS, Bangladeshi HEV sequences clustered with genotype 1a sequences from neighboring countries. Within genotype 1a cluster, Bangladesh HEV strains formed a separate cluster with the 2010 HEV outbreak strains from northern Bangladesh. 80.9 to 100% of the strains had A317T, T735I, L1120I, L1110F, P259S, V1479I, G1634K mutations associates AVH, FHF and RTF. Mutations in T cell recognition epitope T3, T5, T7 was observed in 76.1%, 100% and 100% of the strains respectively. CONCLUSION: Strains of HEV genotype 1a are dominant in Bangladesh and are associated with endemic and outbreak of HEV infection. HEV isolates in Bangladesh have high prevalence of virulence associated mutations and mutation which alters antigenicity to B and T cell epitopes.


Assuntos
Surtos de Doenças , Doenças Endêmicas , Genótipo , Vírus da Hepatite E , Hepatite E , Filogenia , Complicações Infecciosas na Gravidez , Adulto , Bangladesh/epidemiologia , Estudos Transversais , Feminino , Anticorpos Anti-Hepatite/sangue , Hepatite E/sangue , Hepatite E/epidemiologia , Hepatite E/genética , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Humanos , Imunoglobulina M/sangue , Falência Hepática Aguda/sangue , Falência Hepática Aguda/epidemiologia , Falência Hepática Aguda/genética , Masculino , Gravidez , Complicações Infecciosas na Gravidez/epidemiologia , Complicações Infecciosas na Gravidez/genética , Estudos Prospectivos
9.
Biotechnol Lett ; 42(11): 2441-2446, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32875477

RESUMO

OBJECTIVE: Hepatitis E virus (HEV) infection is a major cause of acute hepatitis worldwide. The aim of the study is the development of plant expression system for the production of virus-like particles formed by HEV capsid and the characterization of their immunogenicity. RESULTS: Open reading frame (ORF) 2 encodes the viral capsid protein and possesses candidate for vaccine production. In this study, we used truncated genotype 3 HEV ORF 2 consisting of aa residues 110 to 610. The recombinant protein was expressed in Nicotiana benthamiana plants using the self-replicating potato virus X-based vector pEff up to 10% of the soluble protein fraction. The yield of HEV 110-610 after purification was 150-200 µg per 1 g of green leaf biomass. The recombinant protein formed nanosized virus-like particles. The immunization of mice with plant-produced HEV 110-610 protein induced high levels of HEV-specific serum antibodies. CONCLUSIONS: HEV ORF 2 (110-610 aa) can be used as candidate for the development of a plant-produced vaccine against Hepatitis E.


Assuntos
Vírus da Hepatite E/imunologia , Hepatite E/prevenção & controle , Vacinas contra Influenza/administração & dosagem , Mutação , Proteínas Virais/genética , Animais , Feminino , Anticorpos Anti-Hepatite/sangue , Hepatite E/imunologia , Vírus da Hepatite E/metabolismo , Imunização , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/metabolismo , Injeções Intramusculares , Camundongos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Virais/imunologia
10.
Infect Genet Evol ; 85: 104450, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32629045

RESUMO

Hepatitis E virus (HEV) infection has emerged as an important public health issue. As a zoonotic RNA virus, new strains are continuously discovered from human or various animal species. However, the capability of cross-species infection varies largely among different strains. Because the classical nucleotide-based genotyping system provides little functional insight, this study aimed to comprehensively investigate codon usage of the HEV coding regions for better understanding the evolutional orientation, virus-host interaction and cross-species transmission. We observed significant differences of the four nucleotide usages in the three open reading frames, indicating that the evolutional tendency of HEV caused by mutation pressure is modified by the evolutional dynamic related to positive selection. Furthermore, significant differences of nucleotide usages were found among HEV isolated from different host species, suggesting an important role of natural selection related to the host. Analysis of effective number of codons revealed distinct degrees of biased codon usage caused by mutation pressure or the host. Finally, we have mapped the similarity levels of the overall codon usage between the virus and the host to assess the potential of cross-species infection. Thus, this study has provided a novel aspect for better understanding the HEV genetic orientation and the zoonotic nature.


Assuntos
Uso do Códon , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Hepatite E/virologia , Interações entre Hospedeiro e Microrganismos , Fases de Leitura Aberta , Animais , Composição de Bases , Evolução Molecular , Genoma Viral , Genótipo , Humanos , Mutação , Nucleotídeos/metabolismo , Filogenia , Seleção Genética
11.
Virology ; 544: 1-11, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32174509

RESUMO

The hepatitis E virus (HEV) ORF2 truncated recombinant proteins can self-assemble into virus-like particles (VLPs) and were used as models to investigate the HEV capsid assembly. However, the structural function of the ORF2 C-terminal domain (C52aa from aa 608 to aa 660) remains unclear. Herein, by analyzing a set of ORF2 truncated proteins expressed in Escherichia coli, we found that the highly conserved C-terminal cysteines play a crucial role in the oligomerization of the truncated ORF2 proteins and in their assembly into VLPs, through the formation of dimer-dimer disulfide bonds; and the treatment of native HEV particles with dithiothreitol (DTT) induced the disassembly of the viral capsid, suggesting that the disulfide bonding is required for stabilizing the native HEV capsid. The present study sheds light on the structural role of the C-terminal region of the HEV capsid protein and contributes to the full understating of the viral capsid assembly process.


Assuntos
Vírus da Hepatite E/metabolismo , Proteínas Virais/genética , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Animais , Ditiotreitol/farmacologia , Escherichia coli , Regulação Viral da Expressão Gênica , Vírus da Hepatite E/genética , Proteínas Virais/química
12.
Virology ; 536: 119-124, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31421623

RESUMO

Hepatitis E virus (HEV) causes acute and fulminant hepatitis worldwide. Although enveloped (e) and non-enveloped (ne) forms of HEV have been discovered, host factors involved in infection, including receptors, remain to be elucidated. Here, we identified integrin α3 (encoded by ITGA3), a protein that binds and responds to the extracellular matrix, as an essential host factor for HEV infection. Integrin α3 expression was lower in four HEV-non-permissive cell subclones than in an HEV-permissive subclone. ITGA3 knockout cells lost HEV permissibility, suggesting that integrin α3 is critical for HEV infection. Stable expression of integrin α3 in an HEV-non-permissive subclone provided permissibility only to infection by neHEV; expression of integrin α3 lacking the ectodomain did not. Direct interaction between neHEV and the integrin α3 ectodomain was confirmed by co-precipitation using a soluble integrin α3-Fc. These results strongly suggest that integrin α3 is a key molecule for cellular attachment and entry of neHEV.


Assuntos
Vírus da Hepatite E/genética , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/genética , Integrina alfa3/genética , Internalização do Vírus , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/virologia , Expressão Gênica , Técnicas de Inativação de Genes , Genótipo , Vírus da Hepatite E/metabolismo , Vírus da Hepatite E/patogenicidade , Hepatócitos/patologia , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Integrina alfa3/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Carga Viral , Replicação Viral
13.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31019049

RESUMO

Hepatitis E virus (HEV) is a 7.2-kb positive-sense, single-stranded RNA virus containing three partially overlapping reading frames, ORF1 to ORF3. All nonstructural proteins required for viral replication are encoded by ORF1 and are transcribed as a single transcript. Computational analysis of the complete ORF1 polyprotein identified a previously uncharacterized region of predicted secondary structure bordered by two disordered regions coinciding partially with a region predicted as a putative cysteine protease. Following successful cloning, expression, and purification of this region, the crystal structure of the identified protein was determined and identified to have considerable structural homology to a fatty acid binding domain. Further analysis of the structure revealed a metal binding site, shown unambiguously to specifically bind zinc via a nonclassical, potentially catalytic zinc-binding motif. Based on the structural homology of the HEV protein with known structures, along with the presence of a catalytic zinc-binding motif, it is possible that the identified protein corresponds to the HEV protease, which could require activation or repression through the binding of a fatty acid. This represents a significant step forward in the characterization and the understanding of the molecular mechanisms of the HEV genome. We present analysis for the first time of this identified nonstructural protein, expanding the knowledge and understanding of the complex mechanisms of HEV biology.IMPORTANCE Hepatitis E virus (HEV) is an emerging virus found predominately in developing countries; it causes an estimated 20 million infections, which result in approximately 57,000 deaths a year. Although it is known that the nonstructural proteins of HEV ORF1 are expressed as a single transcript, there is debate as to whether ORF1 functions as a single polyprotein or if it is processed into separate domains via a viral or endogenous cellular protease. Here we present the first structural and biophysical characterization of an HEV nonstructural protein using a construct that has partially overlapping boundaries with the predicted putative cysteine protease.


Assuntos
Proteínas de Transporte/química , Vírus da Hepatite E/metabolismo , Hepatite E/virologia , Proteínas não Estruturais Virais/química , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Cristalografia por Raios X , Vírus da Hepatite E/genética , Humanos , Modelos Moleculares , Fases de Leitura Aberta/genética , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/isolamento & purificação
14.
J Virol ; 93(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30894478

RESUMO

Avian hepatitis E virus (HEV) is the main causative agent of big liver and spleen disease in chickens. Due to the absence of a highly effective cell culture system, there are few reports about the interaction between avian HEV and host cells. In this study, organic anion-transporting polypeptide 1A2 (OATP1A2) from chicken liver cells was identified to interact with ap237, a truncated avian HEV capsid protein spanning amino acids 313 to 549, by a glutathione S-transferase (GST) pulldown assay. GST pulldown and indirect enzyme-linked immunosorbent assays (ELISAs) further confirmed that the extracellular domain of OATP1A2 directly binds with ap237. The expression levels of OATP1A2 in host cells are positively correlated with the amounts of ap237 attachment and virus infection. The distribution of OATP1A2 in different tissues is consistent with avian HEV infection in vivo Finally, when the functions of OATP1A2 in cells are inhibited by its substrates or an inhibitor or blocked by ap237 or anti-OATP1A2 sera, attachment to and infection of host cells by avian HEV are significantly reduced. Collectively, these results displayed for the first time that OATP1A2 interacts with the avian HEV capsid protein and can influence viral infection in host cells. The present study provides new insight to understand the process of avian HEV infection of host cells.IMPORTANCE The process of viral infection is centered around the interaction between the virus and host cells. Due to the lack of a highly effective cell culture system in vitro, there is little understanding about the interaction between avian HEV and its host cells. In this study, a total of seven host proteins were screened in chicken liver cells by a truncated avian HEV capsid protein (ap237) in which the host protein OATP1A2 interacted with ap237. Overexpression of OATP1A2 in the cells can promote ap237 adsorption as well as avian HEV adsorption and infection of the cells. When the function of OATP1A2 in cells was inhibited by substrates or inhibitors, attachment and infection by avian HEV significantly decreased. The distribution of OATP1A2 in different chicken tissues corresponded with that in tissues during avian HEV infection. This is the first finding that OATP1A2 is involved in viral infection of host cells.


Assuntos
Hepevirus/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Animais , Ânions/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Galinhas/virologia , Hepatite E/virologia , Vírus da Hepatite E/metabolismo , Hepatite Viral Animal/virologia , Hepevirus/fisiologia , Peptídeos/metabolismo , Doenças das Aves Domésticas/virologia , Proteínas Virais/metabolismo
15.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30463960

RESUMO

Hepatitis E virus (HEV) is a common cause of acute viral hepatitis worldwide. Most HEV infections are asymptomatic, but immunocompromised patients infected with HEV genotype 3 (HEV3), HEV4, or HEV7 may develop chronic infections. The HEV particles in stools are naked (nHEV), while those in the serum and culture supernatants (eHEV) are associated with lipids. Hepatocytes are polarized epithelial cells that have basolateral (oriented toward the blood) and apical (oriented toward the bile) exosomal pathways. We isolated a subclone, F2, from the human hepatocarcinoma cell line HepG2/C3A that grew as a polarized monolayer culture and had better HEV production than HepG2/C3A cells. F2 cells cultured on semipermeable collagen inserts and infected basolaterally with nHEV3 released 94.6% of virus particles apically, those infected with eHEV3 released 96.8% apically, and eHEV1-infected cells released 99.3% apically. Transcytosis was not involved. Density gradient centrifugation and NP-40 treatment showed that HEV particles released both apically and basolaterally were lipid associated. The apically released HEV3 and HEV1 particles were six and nine times more infectious than those released basolaterally, respectively. Confocal microscopy indicated that the open reading frame 2 (ORF2) capsid protein colocalized apically with ORF3 virus protein, the apical marker DPP4, and the recycling endosome GTPase Rab27a. The amounts of soluble glycosylated ORF2 secreted apically and basolaterally were similar. These polarized-hepatocyte data suggest that infectious HEV particles are mainly released into bile, while the small fraction released into blood could spread HEV throughout the host.IMPORTANCE Hepatitis E virus (HEV) in stools is naked, while that in culture supernatants and patients' blood is lipid associated. Its life cycle in hepatocytes, polarized cells with a basolateral side communicating with blood and an apical side connected with bile, is incompletely understood. We have developed a polarized hepatocyte model and used the cells to analyze the supernatants bathing the apical and basolateral sides and HEV subcellular distribution. HEV particles from both sides were lipid associated, and most infectious HEV particles left the cell via its apical side. Similar amounts of the open reading frame 2 (ORF2) soluble capsid protein were secreted from both sides of the hepatocytes. This model mimicking physiological conditions should help clarify the HEV cell cycle in polarized hepatocytes.


Assuntos
Vírus da Hepatite E/metabolismo , Hepatócitos/virologia , Liberação de Vírus/fisiologia , Proteínas do Capsídeo/metabolismo , Carcinoma Hepatocelular/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Polaridade Celular , Células Epiteliais/virologia , Células Hep G2 , Hepatite E/virologia , Vírus da Hepatite E/patogenicidade , Vírus da Hepatite E/fisiologia , Hepatócitos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Soro/virologia , Proteínas Virais
16.
Artigo em Inglês | MEDLINE | ID: mdl-29530948

RESUMO

Hepatitis E virus (HEV) possesses many of the features of other positive-stranded RNA viruses but also adds HEV-specific nuances, making its virus-host interactions unique. Slow virus replication kinetics and fastidious growth conditions, coupled with the historical lack of an efficient cell culture system to propagate the virus, have left many gaps in our understanding of its structure and replication cycle. Recent advances in culturing selected strains of HEV and resolving the 3D structure of the viral capsid are filling in knowledge gaps, but HEV remains an extremely understudied pathogen. Many steps in the HEV life cycle and many aspects of HEV pathogenesis remain unknown, such as the host and viral factors that determine cross-species infection, the HEV-specific receptor(s) on host cells, what determines HEV chronicity and the ability to replicate in extrahepatic sites, and what regulates processing of the open reading frame 1 (ORF1) nonstructural polyprotein.


Assuntos
Genoma Viral , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Replicação Viral , Vírus da Hepatite E/patogenicidade , Humanos , Vírion , Replicação Viral/fisiologia
17.
DNA Cell Biol ; 37(7): 593-599, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29897788

RESUMO

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.


Assuntos
Antivirais/farmacologia , Vírus da Hepatite E/efeitos dos fármacos , Hepatite E/tratamento farmacológico , Replicação Viral/efeitos dos fármacos , Compostos de Zinco/farmacologia , Animais , Regulação da Expressão Gênica , Hepatite E/genética , Hepatite E/virologia , Vírus da Hepatite E/genética , Vírus da Hepatite E/crescimento & desenvolvimento , Vírus da Hepatite E/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interferons , Interleucinas/genética , Interleucinas/imunologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Transdução de Sinais , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
18.
Sci Rep ; 8(1): 8585, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29872132

RESUMO

Hepatitis E is the most common type of acute hepatitis prevalent worldwide. The open reading frame 3 protein of HEV (HEV ORF3) is proposed to create a favorable environment for viral replication and pathogenesis. However, the mechanisms by which HEV overcomes the effects of host immunity, particularly the role of ORF3, remain to be established. Expression of IFNα and IFNß in supernatant and cell samples was examined via ELISA and quantitative RT-PCR. The protein levels of specific signaling factors in cells overexpressing HEV ORF3 were examined via western blot. Analyses of cells transfected with vectors expressing ORF3 demonstrated that HEV ORF3 significantly impairs the generation of endogenous type I interferon through downregulating TLR3 and TLR7 as well as their corresponding downstream signaling pathways. Moreover, inhibition of NFκB, JAK/STAT and JNK/MAPK signaling pathways contributed significantly to suppression of increased levels of TLR7. Levels of p-P65, p-STAT1 and p-JNK were markedly impaired in ORF3-expressing cells, even upon treatment with the respective agonists. HEV ORF3 inhibits the production of endogenous type I interferon through downregulation of TLR3 and TLR7. Furthermore, suppression of TLR7 is achieved through impairment of multiple signaling pathways, including NFκB, JAK/STAT and JNK/MAPK.


Assuntos
Regulação para Baixo/imunologia , Vírus da Hepatite E/imunologia , Interferon Tipo I/imunologia , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/imunologia , Proteínas Virais/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/genética , Células Hep G2 , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/genética , Células THP-1 , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 3 Toll-Like/metabolismo , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
mBio ; 9(3)2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739903

RESUMO

Approximately 20 million hepatitis E virus (HEV) infections occur annually in both developing and industrialized countries. Most infections are self-limiting, but they can lead to chronic infections and cirrhosis in immunocompromised patients, and death in pregnant women. The mechanisms of HEV replication remain incompletely understood due to scarcity of adequate experimental platforms. HEV undergoes asymmetric genome replication, but it produces an additional subgenomic (SG) RNA encoding the viral capsid and a viroporin in partially overlapping open reading frames. Using a novel transcomplementation system, we mapped the intragenomic subgenomic promoter regulating SG RNA synthesis. This cis-acting element is highly conserved across all eight HEV genotypes, and when the element is mutated, it abrogates particle assembly and release. Our work defines previously unappreciated viral regulatory elements and provides the first in-depth view of the intracellular genome dynamics of this emerging human pathogen.IMPORTANCE HEV is an emerging pathogen causing severe liver disease. The genetic information of HEV is encoded in RNA. The genomic RNA is initially copied into a complementary, antigenomic RNA that is a template for synthesis of more genomic RNA and for so-called subgenomic RNA. In this study, we identified the precise region within the HEV genome at which the synthesis of the subgenomic RNA is initiated. The nucleotides within this region are conserved across genetically distinct variants of HEV, highlighting the general importance of this segment for the virus. To identify this regulatory element, we developed a new experimental system that is a powerful tool with broad utility to mechanistically dissect many other poorly understood functional elements of HEV.


Assuntos
Vírus da Hepatite E/genética , Hepatite E/virologia , Regiões Promotoras Genéticas , RNA Viral/genética , Sequência de Bases , Regulação Viral da Expressão Gênica , Genoma Viral , Vírus da Hepatite E/metabolismo , Humanos , Dados de Sequência Molecular , RNA Viral/metabolismo , Transcrição Gênica
20.
Liver Int ; 38(2): 266-277, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28718943

RESUMO

BACKGROUND AND AIMS: Hepatitis E virus (HEV) is a major cause of acute viral hepatitis with >3 million symptomatic cases per year accounting for 70 000 HEV-related deaths. HEV-specific T-cell responses have been investigated against structural proteins expressed by open reading frames (ORF) 2 and 3. T-cell responses against non-structural HEV proteins encoded by ORF1 are hardly studied. The aim of this study was to determine HEV ORF1-specific T-cell responses in comparison to ORF2/3 in patients exposed to HEV. METHODS: HEV-specific CD4+ and CD8+ T-cell responses against HEV genotype 3 were investigated in patients with acute and chronic hepatitis E as well as in HEV seropositive and seronegative individuals. HEV-specific T-cell responses were determined by proliferation and intracellular cytokine assay upon stimulation of PBMCs with HEV-specific overlapping peptide pools spanning the entire HEV genome. HEV-antigen was measured using an anti-HEV antigen-specific ELISA. RESULTS: Broad HEV ORF1-specific T-cell responses were detected in patients with acute, resolved and chronic hepatitis E without distinct dominant regions. The magnitude and frequency in recognition of ORF1-specific T-cell responses were similar compared to responses against HEV ORF2/3. Longitudinal studies of HEV-specific T-cell responses displayed similar behaviour against structural and non-structural proteins. HEV-antigen levels were inversely correlated with HEV-specific T-cell responses. CONCLUSIONS: HEV-specific T-cell responses are detectable against the entire HEV genome including the non-structural proteins. HEV-specific T-cell responses are associated with control of HEV infection. These findings have implications for the design of HEV vaccines.


Assuntos
Proliferação de Células , Vírus da Hepatite E/imunologia , Hepatite E/imunologia , Hepatite Autoimune/imunologia , Ativação Linfocitária , Fases de Leitura Aberta , Linfócitos T/imunologia , Proteínas Virais/imunologia , Doença Aguda , Adolescente , Adulto , Idoso , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Genótipo , Hepatite E/diagnóstico , Hepatite E/virologia , Vírus da Hepatite E/genética , Vírus da Hepatite E/metabolismo , Hepatite Autoimune/diagnóstico , Hepatite Autoimune/metabolismo , Hepatite Autoimune/virologia , Interações Hospedeiro-Patógeno , Humanos , Masculino , Pessoa de Meia-Idade , Linfócitos T/metabolismo , Linfócitos T/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...