Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virology ; 595: 110091, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38718446

RESUMO

Preliminary investigations have demonstrated that the cysteines located at the C-terminus of HEV ORF2 protein exhibits disulfide bonding capability during virus-like particles (VLPs) assembly. However, the effect and mechanism underlying the pairing of disulfide bonds formed by C627, C630, and C638 remains unclear. The p222 protein encompasses C-terminus and serves as a representative of HEV ORF2 to investigate the specific impacts of C627, C630, and C638. The three cysteines were subjected to site-directed mutagenesis and expressed in prokaryotes; Both the mutated proteins and p222 underwent polymerization except for p222A; Surprisingly, only p222 was observed as abundant spherical particles under transmission electron microscope (TEM); Stability and immunogenicity of the p222 exhibited higher than other mutated proteins; LC/MS/MS analysis identified four disulfide bonds in the p222. The novel findings suggest that the three cysteines contribute to structural and functional properties of ORF2 protein, highlighting the indispensability of each cysteine.


Assuntos
Cisteína , Vírus da Hepatite E , Proteínas Virais , Cisteína/química , Cisteína/metabolismo , Vírus da Hepatite E/genética , Vírus da Hepatite E/química , Proteínas Virais/genética , Proteínas Virais/química , Proteínas Virais/metabolismo , Mutagênese Sítio-Dirigida , Dissulfetos/química , Dissulfetos/metabolismo , Animais , Humanos
2.
Protein Expr Purif ; 187: 105947, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34314826

RESUMO

Hepatitis E virus (HEV) is a major causative agent of hepatitis E infections across the globe. Although the essentiality of HEV nonstructural polyprotein (pORF1) putative Y-domain (Yd) has been established in viral pathogenesis, its structural-functional role remains elusive. The current research discusses the novel exploration on Yd protein expression, purification, biophysical characterization and structure-based docking analysis. The codon optimized synthetic gene and optimized expression parameters i.e., 5 h induction with 0.25 mM IPTG at 37 °C, resulted in efficient production of Yd protein (~40 kDa) in E. coli BL21(DE3) cells. Majority of the recombinant Yd (rYd) protein expressed as inclusion bodies was solubilized in 0.5% N-lauroylsarcosine and purified using Ni-NTA chromatography. Circular dichroism (CD) and UV visible absorption spectroscopic studies on Yd revealed both secondary and tertiary structure stability in alkaline range (pH 8.0-10.0), suggesting correlation with its physiological activity. Thus, loss in structure at low pH perhaps play crucial role in cytoplasmic-membrane interaction. The biophysical data were in good agreement with insilico structural analyses, which suggested mixed α/ß fold, non-random and basic nature of Yd protein. Furthermore, due to Yd protein essentiality in HEV replication and pathogenesis, it was considered as a template for docking and drug-likeness analyses. The 3D modeling of Yd protein and structure-based screening and drug-likeness of inhibitory compounds, including established antiviral drugs led to the identification of top nine promising candidates. Nonetheless, in vitro studies on the predicted interaction of Yd with intracellular-membrane towards establishing replication-complexes as well as validations of the proposed therapeutic agents are warranted.


Assuntos
Vírus da Hepatite E/química , Proteínas Recombinantes/química , Proteínas Virais/química , Sequência de Aminoácidos , Domínio Catalítico , Escherichia coli/genética , Vírus da Hepatite E/genética , Humanos , Corpos de Inclusão/química , Simulação de Acoplamento Molecular , Conformação Proteica , Proteínas Recombinantes/genética , Solubilidade , Proteínas Virais/genética
3.
Antiviral Res ; 185: 104997, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33326835

RESUMO

Hepatitis E virus (HEV) causes 14 million infections and 60,000 deaths per year globally, with immunocompromised persons and pregnant women experiencing severe symptoms. Although ribavirin can be used to treat chronic hepatitis E, toxicity in pregnant patients and the emergence of resistant strains are major concerns. Therefore there is an imminent need for effective HEV antiviral agents. The aims of this study were to develop a drug screening platform and to discover novel approaches to targeting steps within the viral life cycle. We developed a screening platform for molecules inhibiting HEV replication and selected a candidate, isocotoin. Isocotoin inhibits HEV replication through interference with heat shock protein 90 (HSP90), a host factor not previously known to be involved in HEV replication. Additional work is required to understand the compound's translational potential, however this suggests that HSP90-modulating molecules, which are in clinical development as anti-cancer agents, may be promising therapies against HEV.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Vírus da Hepatite E/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Antivirais/isolamento & purificação , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Proteínas de Choque Térmico HSP90/metabolismo , Hepatite E/tratamento farmacológico , Vírus da Hepatite E/química , Humanos , Ligação Proteica , Replicação Viral/efeitos dos fármacos
4.
Biochem Biophys Res Commun ; 532(1): 25-31, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-32819713

RESUMO

Hepatitis E virus (HEV) is a non-enveloped, globular particle that is responsible for acute hepatitis. HEV is classified into the Hepeviridae family and can be divided into four species (A-D). All HEV variants that infect humans are reported to belong to species A (HEV-A), except species C (HEV-C), which was reported to infect humans in December 2018. We determined the crystal structure of the HEV-C E2s domain at 1.8 Å resolution. It contains a classical 12-stranded ß-sandwich motif and forms dimers by hydrogen bonding, though the amino acid residues that form hydrogen bonds are quite different from the residues of HEV-A. The HEV-C E2s domain shares the common groove region with other structurally related viruses, and some subtle differences in this region may be related to host adoption or antibody binding. Antibody binding experiments and structural analysis revealed that HEV-C E2s is able to bind to the previously reported broad-spectrum antibody 8G12 but not bind to the antibody 8C11. Meanwhile, the structure analysis shows that HEV-C E2s does not have the key sites for binding to host cells as displayed by HEV-A (Genotype 1) E2s. These structural and biological findings present important implications for understanding the molecular mechanisms of host recognition and entry of HEV-C, as well as provide clues to the development of therapeutic antibodies and vaccines against HEV-C infection.


Assuntos
Vírus da Hepatite E/química , Proteínas do Envelope Viral/química , Anticorpos Antivirais , Antígenos Virais/química , Antígenos Virais/genética , Cristalografia por Raios X , Epitopos/química , Epitopos/genética , Hepatite E/imunologia , Hepatite E/virologia , Vírus da Hepatite E/genética , Vírus da Hepatite E/fisiologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformação Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia
5.
J Vis Exp ; (135)2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29806824

RESUMO

Virus-like particles (VLPs) have been used as nanocarriers to display foreign epitopes and/or deliver small molecules in the detection and treatment of various diseases. This application relies on genetic modification, self-assembly, and cysteine conjugation to fulfill the tumor-targeting application of recombinant VLPs. Compared with genetic modification alone, chemical conjugation of foreign peptides to VLPs offers a significant advantage because it allows a variety of entities, such as synthetic peptides or oligosaccharides, to be conjugated to the surface of VLPs in a modulated and flexible manner without alteration of the VLP assembly. Here, we demonstrate how to use the hepatitis E virus nanoparticle (HEVNP), a modularized theranostic capsule, as a multifunctional delivery carrier. Functions of HEVNPs include tissue-targeting, imaging, and therapeutic delivery. Based on the well-established structural research of HEVNP, the structurally independent and surface-exposed residues were selected for cysteine replacement as conjugation sites for maleimide-linked chemical groups via thiol-selective linkages. One particular cysteine-modified HEVNP (a Cys replacement of the asparagine at 573 aa (HEVNP-573C)) was conjugated to a breast cancer cell-specific ligand, LXY30 and labeled with near-infrared (NIR) fluorescence dye (Cy5.5), rendering the tumor-targeted HEVNPs as effective diagnostic capsules (LXY30-HEVNP-Cy5.5). Similar engineering strategies can be employed with other macromolecular complexes with well-known atomic structures to explore potential applications in theranostic delivery.


Assuntos
Vírus da Hepatite E/química , Nanopartículas/química , Animais , Humanos , Insetos/virologia
6.
Nat Rev Gastroenterol Hepatol ; 15(2): 96-110, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29162935

RESUMO

At least 20 million hepatitis E virus (HEV) infections occur annually, with >3 million symptomatic cases and ∼60,000 fatalities. Hepatitis E is generally self-limiting, with a case fatality rate of 0.5-3% in young adults. However, it can cause up to 30% mortality in pregnant women in the third trimester and can become chronic in immunocompromised individuals, such as those receiving organ transplants or chemotherapy and individuals with HIV infection. HEV is transmitted primarily via the faecal-oral route and was previously thought to be a public health concern only in developing countries. It is now also being frequently reported in industrialized countries, where it is transmitted zoonotically or through organ transplantation or blood transfusions. Although a vaccine for HEV has been developed, it is only licensed in China. Additionally, no effective, non-teratogenic and specific treatments against HEV infections are currently available. Although progress has been made in characterizing HEV biology, the scarcity of adequate experimental platforms has hampered further research. In this Review, we focus on providing an update on the HEV life cycle. We will further discuss existing cell culture and animal models and highlight platforms that have proven to be useful and/or are emerging for studying other hepatotropic (viral) pathogens.


Assuntos
Hepatite E/epidemiologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Saúde Global , Hepatite E/terapia , Hepatite E/transmissão , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Vírus da Hepatite E/fisiologia , Hepatócitos/virologia , Células-Tronco Pluripotentes/virologia , Vacinas contra Hepatite Viral , Proteínas Virais/fisiologia , Tropismo Viral/fisiologia , Vírion/química , Vírion/fisiologia
7.
Virus Res ; 208: 98-109, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26087402

RESUMO

Lack of robust cell culture systems for Hepatitis E virus (HEV) infection has hampered understanding of HEV biology. We attempted to identify the host cellular factors that interact with HEV 5' and 3' untranslated regions (UTRs) by RNA affinity chromatography followed by mass spectrometry analysis. Hepatitis E virus genotype-1 (HEV-1) and Hepatitis E virus genotype-4 (HEV-4) and three cell lines (HepG2/C3A, A549 and Caco2) were employed to understand the UTR-host protein interaction. RNA pull-down and matrix-assisted laser desorption/ionization time-of-flight/time-of-flight (MALDI TOF/TOF) analysis revealed that DHX9, PTK-7, DIS3 and TCR E chain (CD3ɛ) of all the three cell lines interacted with HEV 3'UTR while RAD50 and TLE-4 interacted with HEV 5'UTR. RNA immuno-precipitation studies further confirmed the interaction of DHX9, DIS3 and TCR E chain. The expression changes in genes associated with the identified proteins were quantitated in the peripheral blood mononuclear cells (PBMCs) of Hepatitis E patients during acute and recovery phases. The data revealed that HEV infection influences the exosomes, T cell receptor signalling and Wnt signalling pathways. Interactions of DIS3 with HEV UTRs suggest that exosomes might have important implication in HEV life cycle.


Assuntos
Vírus da Hepatite E/metabolismo , Hepatite E/metabolismo , Regiões não Traduzidas , Células CACO-2 , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Hepatite E/genética , Hepatite E/virologia , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Humanos , Leucócitos Mononucleares/química , Leucócitos Mononucleares/metabolismo , Espectrometria de Massas , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo
8.
J Virol ; 88(20): 12017-25, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100839

RESUMO

Little is known about virus adaptation in immunocompromised patients with chronic genotype 3 hepatitis E virus (HEV3) infections. Virus-host recombinant strains have been isolated recently from chronically infected patients. The nature and incidence of such recombinant events occurring during infections of solid-organ transplant (SOT) recipients are essentially unknown. The polyproline region (PPR) of strains isolated from SOT patients was sequenced during the acute-infection phase (n = 59) and during follow-up of patients whose infections became chronic (n = 27). These 27 HEV strains included 3 (11%) that showed recombinant events 12, 34, 48, or 88 months after infection. In one strain, parts of the PPR and the RNA-dependent RNA polymerase were concomitantly inserted. In the second, a fragment of a human tyrosine aminotransferase (TAT) gene was inserted first, followed by a fragment of PPR. A fragment of the human inter-α-trypsin inhibitor (ITI) gene was inserted in the third. All the inserted sequences were rich in aliphatic and basic amino acids. In vitro growth experiments suggest that the ITI insertion promoted more vigorous virus growth. In silico studies showed that the inserted sequences could provide potential acetylation, ubiquitination, and phosphorylation sites. We found that recombinant events had occurred in the HEV PPR in approximately 11% of the strains isolated from chronically infected transplant patients followed up in Toulouse University Hospital. These inserted fragments came from the HEV genome or a human gene and could enhance virus replication. Importance: Hepatitis E virus (HEV) can cause chronic infections in immunocompromised patients, including solid-organ transplant (SOT) recipients. Two strains that had undergone recombination with human ribosomal genes were described recently. The strains with inserted sequences replicated better in vitro. Little is known about the frequency of such recombinant events or how such an insertion enhances replication. We therefore investigated 59 SOT patients infected with HEV and found 3 strains with 4 recombinant events in 27 of these patients whose infection became chronic. The 4 inserted sequences were of different origins (human gene or HEV genome), but all were enriched in aliphatic and basic amino acids and provided potential regulation sites. Our data indicate that recombinant events occur in approximately 11% of strains isolated from chronically infected patients. The structures of the inserted sequences provide new clues as to how the inserted sequences could foster virus replication.


Assuntos
Vírus da Hepatite E/química , Hospedeiro Imunocomprometido , Transplante de Órgãos , Peptídeos/química , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Genoma Viral , Vírus da Hepatite E/genética , Vírus da Hepatite E/patogenicidade , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos
9.
Virus Res ; 179: 26-33, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24333153

RESUMO

Hepatitis E virus (HEV), the causative agent of hepatitis E, is a non-enveloped RNA virus. The open reading frame 1 encoded non-structural polyprotein has putative domains for methyltransferase, cysteine protease, helicase and RNA-dependent RNA polymerase, however processing of this polyprotein is still uncertain. HEV helicase belongs to superfamily 1 and has all seven conserved motifs typical of the family. NTPase and RNA duplex unwinding activities of HEV helicase domain were recently demonstrated by us. A non-radioactive RNA unwinding assay was developed using biotin and digoxigenin labeled duplex RNA substrate with 5' overhangs for measuring strand displacement activity of the helicase. A series of deletion mutants were constructed to investigate role of individual motifs in the enzymatic activities. Deletion mutants for motif M I and M IV showed increase in ATPase activity. Deletion mutant M VI retained ATPase activity comparable to wild type protein. Mutant M II showed reduced ATPase activity (P=0.003) with no significant decrease in unwinding activity while mutants M Ia and M III showed major reduction of both ATPase and unwinding activities indicating crucial role of these motifs in the helicase function. Overall analysis of deletion mutants showed that Motif I, IV, V and VI have alternative motifs to carry out enzymatic functions of the protein while motifs Ia and III are critical as well as unique motifs in the protein. Knowing the important role of helicase protein during positive sense RNA virus replication, these unique motifs could be good antiviral targets.


Assuntos
Vírus da Hepatite E/enzimologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Mutagênese , Mutação , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Virais/química
10.
Virus Res ; 179: 220-4, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24321124

RESUMO

The biochemical or biophysical characterization of a papain-like cysteine protease in HEV ORF1-encoded polyprotein still remains elusive. Very recently, we have demonstrated the indispensability of ORF1 protease-domain cysteines and histidines in HEV replication, ex vivo (Parvez, 2013). In this report, the polyprotein partial sequences of HEV strains and genetically-related RNA viruses were analyzed, in silico. Employing the consensus-prediction results of RUBV-p(150) protease as structural-template, a 3D model of HEV-protease was deduced. Similar to RUBV-p(150), a 'papain-like ß-barrel fold' structurally confirmed the classification of HEV-protease. Further, we recognized a catalytic 'Cys434-His443' dyad homologue of RUBV-p(150) (Cys1152-His1273) and FMDV-L(pro) (Cys51-His148) in line with our previous mutational analysis that showed essentiality of 'His443' but not 'His590' in HEV viability. Moreover, a RUBV 'Zn(2+) binding motif' (Cys1167-Cys1175-Cys1178-Cys1225-Cys1227) equivalent of HEV was identified as 'Cys457-His458-Cys459 and Cys481-Cys483' residues within the 'ß-barrel fold'. Notably, unlike RUBV, 'His458' also clustered therein, that was in conformity with the consensus cysteine protease 'Zn(2+)-binding motif'. By homology, we also proposed an overlapping 'Ca(2+)-binding site' 'D-X-[DNS]-[ILVFYW]-[DEN]-G-[GP]-XX-DE' signature, and a 'proline-rich motif' interacting 'tryptophan (W437-W472)' module in the modeled structure. Our analysis of the predicted model therefore, warrants critical roles of the 'catalytic dyad' and 'divalent metal-binding motifs' in HEV protease structural-integrity, ORF1 self-processing, and RNA replication. This however, needs further experimental validations.


Assuntos
Cisteína Proteases/química , Vírus da Hepatite E/enzimologia , Proteínas Virais/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Cisteína Proteases/genética , Cisteína Proteases/metabolismo , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Modelos Moleculares , Dados de Sequência Molecular , Fases de Leitura Aberta , Estrutura Secundária de Proteína , Alinhamento de Sequência , Proteínas Virais/genética , Proteínas Virais/metabolismo , Zinco/química , Zinco/metabolismo
11.
J Virol ; 86(18): 10186-93, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22811526

RESUMO

The hepatitis E virus (HEV) polyproline region (PPR) is an intrinsically unstructured region (IDR). This relaxed structure allows IDRs, which are implicated in the regulation of transcription and translation, to bind multiple ligands. Originally the nucleotide variability seen in the HEV PPR was assumed to be due to high rates of insertion and deletion. This study shows that the mutation rate is about the same in the PPR as in the rest of the nonstructural polyprotein. The difference between the PPR and the rest of the polyprotein is due to the higher tolerance of the PPR for substitutions at the first and second codon positions. With this higher promiscuity there is a shift in nucleotide occupation of these codons leading to translation of more cytosine residues: a shift that leads to more proline, alanine, serine, and threonine being encoded rather than histidine, phenylalanine, tryptophan, and tyrosine. This pattern of amino acid usage is typical of proline-rich IDRs. Increased usage of cytosine also leads to >22% of all amino acids in the PPR being prolines. Alignments of PPR sequences from HEV strains representing all genotypes indicate that all zoonotic isolates share an ancestor, and the carboxyl half of the PPR is more tolerant of mutations than the amino half. The evolution of HEV PPR, in contrast with that of the rest of the nonstructural polyprotein, is molded by pressures that lead toward increased proline usage with a corresponding decrease in the usage of aromatic amino acids, favoring formation of IDR structures.


Assuntos
Evolução Molecular , Vírus da Hepatite E/genética , Peptídeos/genética , Proteínas não Estruturais Virais/genética , Animais , Sequência de Bases , Códon/genética , Variação Genética , Vírus da Hepatite E/química , Vírus da Hepatite E/classificação , Humanos , Mutação INDEL , Dados de Sequência Molecular , Mutação , Peptídeos/química , RNA Viral/genética , Coelhos , Homologia de Sequência do Ácido Nucleico , Proteínas não Estruturais Virais/química
12.
PLoS One ; 7(4): e35974, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22545153

RESUMO

Genomes of hepatitis E virus (HEV), rubivirus and cutthroat virus (CTV) contain a region of high proline density and low amino acid (aa) complexity, named the polyproline region (PPR). In HEV genotypes 1, 3 and 4, it is the only region within the non-structural open reading frame (ORF1) with positive selection (4-10 codons with dN/dS>1). This region has the highest density of sites with homoplasy values >0.5. Genotypes 3 and 4 show ∼3-fold increase in homoplastic density (HD) in the PPR compared to any other region in ORF1, genotype 1 does not exhibit significant HD (p<0.0001). PPR sequence divergence was found to be 2-fold greater for HEV genotypes 3 and 4 than for genotype 1. The data suggest the PPR plays an important role in host-range adaptation. Although the PPR appears to be hypervariable and homoplastic, it retains as much phylogenetic signal as any other similar sized region in the ORF1, indicating that convergent evolution operates within the major HEV phylogenetic lineages. Analyses of sequence-based secondary structure and the tertiary structure identify PPR as an intrinsically disordered region (IDR), implicating its role in regulation of replication. The identified propensity for the disorder-to-order state transitions indicates the PPR is involved in protein-protein interactions. Furthermore, the PPR of all four HEV genotypes contains seven putative linear binding motifs for ligands involved in the regulation of a wide number of cellular signaling processes. Structure-based analysis of possible molecular functions of these motifs showed the PPR is prone to bind a wide variety of ligands. Collectively, these data suggest a role for the PPR in HEV adaptation. Particularly as an IDR, the PPR likely contributes to fine tuning of viral replication through protein-protein interactions and should be considered as a target for development of novel anti-viral drugs.


Assuntos
Vírus da Hepatite E/genética , Hepatite E/virologia , Peptídeos/genética , Proteínas Virais/genética , Adaptação Fisiológica , Sequência de Aminoácidos , Genótipo , Vírus da Hepatite E/química , Vírus da Hepatite E/fisiologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fases de Leitura Aberta , Peptídeos/química , Filogenia , Alinhamento de Sequência , Proteínas Virais/química
13.
J Virol ; 86(10): 5637-46, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438540

RESUMO

The ORF3 protein of hepatitis E virus (HEV) is a multifunctional protein important for virus replication. The ORF3 proteins from human, swine, and avian strains of HEV contain a conserved PXXP amino acid motif, resembling either Src homology 3 (SH3) cell signaling interaction motifs or "late domains" involved in host cell interactions aiding in particle release. Using an avian strain of HEV, we determined the roles of the conserved prolines within the PREPSAPP motif in HEV replication and infectivity in Leghorn male hepatoma (LMH) chicken liver cells and in chickens. Each proline was changed to alanine to produce 8 avian HEV mutants containing single mutations (P64, P67, P70, and P71 to A), double mutations (P64/67A, P64/70A, and P67/70A), and triple mutations (P64/67/70A). The results showed that avian HEV mutants are replication competent in vitro, and none of the prolines in the PXXPXXPP motif are essential for infectivity in vivo; however, the second and third prolines appear to aid in fecal virus shedding, suggesting that the PSAP motif, but not the PREP motif, is involved in virus release. We also showed that the PSAP motif interacts with the host protein tumor suppressor gene 101 (TSG101) and that altering any proline within the PSAP motif disrupts this interaction. However, we showed that the ORF2 protein expressed in LMH cells is efficiently released from the cells in the absence of ORF3 and that coexpression of ORF2 and ORF3 did not act synergistically in this release, suggesting that another factor(s) such as ORF1 or viral genomic RNA may be necessary for proper particle release.


Assuntos
Hepatite E/virologia , Hepevirus/fisiologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Liberação de Vírus , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Galinhas , Modelos Animais de Doenças , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Vírus da Hepatite E/fisiologia , Hepevirus/química , Hepevirus/genética , Humanos , Masculino , Dados de Sequência Molecular , Fases de Leitura Aberta , Proteínas Virais/genética , Replicação Viral
14.
Virol J ; 6: 186, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19887014

RESUMO

BACKGROUND: Swine hepatitis E virus (swHEV) is a zoonotic disease that is considered a major problem in pig production and presents a threat to human health. Elucidation of the major antigenic epitopes of swHEV is essential for the effective control of swHEV epidemics. RESULTS: By bioinformatic analysis, we identified and then synthesized 12 peptides from open reading frames (ORFs) ORF1, ORF2 and ORF3, including swHEV-1 - swHEV-12. Using the results from ELISA, we selected swHEV-11 as the best candidate antigen and used it as a coating antigen for the development of peptide-based swine anti-HEV ELISA kits. The coefficient of variation (CV) the coefficient of variation (CV) varied between 4.3-7.2% in the same batch, and between 8.2-17.7% in six different batches. When comparing our swine peptide-based kit with the commercial recombinant-based kit, the humane anti-HEV IgG test had a 73.4% correspondence rate for them. CONCLUSION: This is the first systemic study to screen the diagnostic peptides of swHEV and our findings strongly suggest that peptide swHEV-11 is a potent diagnostic reagent of swHEV that could be used in the development of highly efficient diagnostic assays for the specific and highly sensitive detection of anti-HEV activity in swine serum samples.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Antígenos de Hepatite/sangue , Vírus da Hepatite E/imunologia , Hepatite E/veterinária , Peptídeos/sangue , Doenças dos Suínos/sangue , Doenças dos Suínos/diagnóstico , Sequência de Aminoácidos , Animais , Antígenos de Hepatite/química , Antígenos de Hepatite/imunologia , Hepatite E/diagnóstico , Hepatite E/imunologia , Vírus da Hepatite E/química , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Peptídeos/química , Peptídeos/imunologia , Estrutura Secundária de Proteína , Reprodutibilidade dos Testes , Suínos , Doenças dos Suínos/imunologia
15.
J Virol ; 80(17): 8493-502, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16912299

RESUMO

Macro domains constitute a protein module family found associated with specific histones and proteins involved in chromatin metabolism. In addition, a small number of animal RNA viruses, such as corona- and toroviruses, alphaviruses, and hepatitis E virus, encode macro domains for which, however, structural and functional information is extremely limited. Here, we characterized the macro domains from hepatitis E virus, Semliki Forest virus, and severe acute respiratory syndrome coronavirus (SARS-CoV). The crystal structure of the SARS-CoV macro domain was determined at 1.8-Angstroms resolution in complex with ADP-ribose. Information derived from structural, mutational, and sequence analyses suggests a close phylogenetic and, most probably, functional relationship between viral and cellular macro domain homologs. The data revealed that viral macro domains have relatively poor ADP-ribose 1"-phosphohydrolase activities (which were previously proposed to be their biologically relevant function) but bind efficiently free and poly(ADP-ribose) polymerase 1-bound poly(ADP-ribose) in vitro. Collectively, these results suggest to further evaluate the role of viral macro domains in host response to viral infection.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Vírus da Hepatite E/química , Histonas/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Vírus da Floresta de Semliki/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Adenosina Difosfato Ribose/análogos & derivados , Adenosina Difosfato Ribose/química , Sequência de Aminoácidos , Cristalografia , Vírus da Hepatite E/metabolismo , Histonas/química , Modelos Moleculares , Dados de Sequência Molecular , Poli(ADP-Ribose) Polimerases/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Vírus da Floresta de Semliki/metabolismo , Relação Estrutura-Atividade
16.
Sheng Wu Gong Cheng Xue Bao ; 20(1): 90-8, 2004 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-16108497

RESUMO

Hepatitis E is a main cause of acute viral hepatitis in developing countries where it occurs as sporadic cases and in epidemics form. The causative agent, hepatitis E virus, is transmitted primarily by the fecal-oral route. The approximately 7.5 kb positive-sense single-strand RNA genome includes three open reading frames (ORFs), one of which (ORF2) is postulated to encode the major viral capsid protein (pORF2) of 660 amino acid residues. We earlier showed that a bacterially expressed peptide, designated as NE2, located from amino acid residues 394 to 606 of ORF2, was found to aggregate into homodimer to at least hexamer. To understand the interface domains within this peptide vital for dimerization and formation of major neutralizing epitopes, NE2 protein underwent terminal-truncated and site-directed mutation. The hydrophobic region, ORF2 aa597-aa602 (AVAVLA), played a key role in oligomerization. Any amino acid residue of this region replaced with glutamic acid residue, the peptide can not refold as homodimer and/or oligomer. The immunoreactivities of these mutant peptides, blotted with anti-HEV neutralizing monoclonal antibody (8C11) and convalescent human sera, show associated to the formation of homodimer. The intermolecular contact region on homodimer was investigated by chemical cross-linking of two site-directed cysteines. When the alanine on aa597 site mutated with cysteine, two different homodimers were found in SDS-PAGE analysis. One (42kD) can be disassociated with 8mol/L urea, which is postulated to form by virtue of hydrophobic interaction, and the other (60kD) falls apart with the reductant DTT present. The exact conformation, generating the cross-linking reaction of cysteines, was further investigated by induced-oxidation on monomer and hydrophobic homodimer of A597C protein with GSH/GSSG. And the results revealed, it is the conformation of hydrophobic homodimer that induces the disulfide bond come into being, instead of the one of monomer. So the aa597 site was verified to be located on interface domain of hydrophobically interacting homodimeric complex. To evaluate the biological significance of hydrophobicity of interface domain, we searched natural variations as to the region on all available databases with NCBI blast program. All variations on these amino acid residues kept higher hydrophobicity, which suggests that the hydrophobic domain is critical for the assemblage and propagation of HEV. NE2 N-terminal deletions up to aa458 had no effect on dimerization and took no exact part in formation of major neutralizing epitopes, but the fragment may act as helper for the formation of major neutralizing epitopes on NE2. Interestingly, the C-terminus aa605-aa660 of ORF2 can also act as helper instead of the N-terminus of NE2. This study suggests an interface domain of NE2 might be vital for HEV capsomer assembly and formation of major neutralizing epitopes. These results may offer clues to the rational design of recombinant anti-HEV vaccine.


Assuntos
Proteínas do Capsídeo/química , Vírus da Hepatite E/química , Multimerização Proteica , Interações Hidrofóbicas e Hidrofílicas , Estrutura Terciária de Proteína , Montagem de Vírus
17.
J Med Virol ; 64(2): 125-32, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11360244

RESUMO

A 23 kDa peptide locating to amino acid residues 394 to 604 of the major Hepatitis E Virus (HEV) structural protein was expressed in E. coli. This peptide was found to interact naturally with one another to form homodimers and it was recognized strongly and commonly in its dimeric form by HEV reactive human sera. The antigenic activity associated with the dimeric form was abrogated when the dimer was dissociated into monomer and the activity was reconstituted after the monomer was re-associated into dimer again. The dimeric form of the peptide elicited a vigorous antibody response in experimental animals and the resulting antisera were found to cross-react against HEV, effecting an efficient immune capture of the virus. These results attributed the antigenic activity associated with the dimeric form of the peptide to conformational antigenic determinants generated as a result of interaction between the peptide molecules. It is suggested that some of these antigenic determinants may be expressed by the HEV capsid and raised the possibility of this bacterially expressed peptide as an HEV vaccine candidate.


Assuntos
Vírus da Hepatite E/química , Hepatite E/virologia , Proteínas Estruturais Virais/genética , Animais , Western Blotting , Epitopos/biossíntese , Epitopos/química , Epitopos/imunologia , Escherichia coli/genética , Anticorpos Anti-Hepatite/sangue , Antígenos de Hepatite/biossíntese , Antígenos de Hepatite/química , Antígenos de Hepatite/imunologia , Hepatite E/sangue , Humanos , Soros Imunes/imunologia , Fases de Leitura Aberta , Coelhos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/imunologia
18.
Zhonghua Yu Fang Yi Xue Za Zhi ; 35(5): 315-7, 2001 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-11769630

RESUMO

OBJECTIVE: To develop a diagnostic kit for detecting serum anti-hepatitis E virus (HEV) IgG with enzyme-linked immunoassay (ELISA). METHODS: The diagnostic kit of detecting anti-HEV IgG with ELISA was prepared by two synthetic HEV peptides used for coating the solid-phase to capture the antibody against HEV in serum, and then by horseradish peroxidase antihuman IgG (gamma chain) added as the second antibody to bind the anti-HEV IgG in serum. Its sensitivity, specificity, precision and stability were measured and clinically evaluated. RESULTS: The sensitivity, specificity, and precision of the diagnostic kit detected by the China National Institute for the Control of Pharmaceutical and Biological Products were 90% (1/10), 100% (0/30) and < 15%, respectively, meeting the national standards for detecting serum anti-HEV IgG with ELISA. The ELISA Kit is stable for 1 year at least under 4 degrees C. The total consistency rates were 100% (43/43) and 96.6% (86/89), as compared with Genelabs and Singaporean DBL kits. This diagnostic kit for detecting serum anti-HEV IgG with ELISA was evaluated and approved by the State Drug Administration, China and a certificate was conferred in 1998. CONCLUSIONS: The diagnostic kit for detecting anti-HEV IgG with ELISA can be used for epidemiological studies and clinical diagnosis of HEV infection.


Assuntos
Vírus da Hepatite E/imunologia , Hepatite E/diagnóstico , Imunoglobulina G/sangue , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática/normas , Vírus da Hepatite E/química , Humanos , Peptídeos/síntese química , Peptídeos/imunologia , Kit de Reagentes para Diagnóstico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
19.
Vopr Virusol ; 44(2): 60-4, 1999.
Artigo em Russo | MEDLINE | ID: mdl-10358898

RESUMO

Antigenic specificity of recombinant polypeptides HE40 and HE60 containing fragments of gene ORF2 and ORF3 protein products of hepatitis E, strain Burma, produced in E. coli cells, is analyzed. Blood sera from patients with acute hepatitis from an endemic region in Uzbekistan were tested for IgG to recombinant antigens by solid-phase enzyme immunoassay with a protein fragment coded by PRF3 gene, a synthetic peptide previously characterized in a commercial test system, as the positive control. 93% sera reacting with recombinant polypeptide HE60 and 32% reacting with HE40 reacted with the synthetic peptide. No antibodies to the studied polypeptides were detected in the sera of Moscow patients with hepatitis A, B, or C confirmed by laboratory findings. Antigenic specificity of recombinant polypeptide HE60 was confirmed by competitive enzyme immunoassay with the same peptide as the competitive antigen. Test system based on recombinant polypeptides HE40 and HE60 was used for deciphering the etiological structure of acute viral hepatitis which occurred in a hepatitis E endemic region of Uzbekistan in 1993.


Assuntos
Reações Antígeno-Anticorpo , Antígenos Virais/imunologia , Vírus da Hepatite E/química , Fragmentos de Peptídeos/imunologia , Proteínas Virais/química , Sequência de Aminoácidos , Hepatite Viral Humana/sangue , Hepatite Viral Humana/imunologia , Humanos , Técnicas Imunoenzimáticas , Dados de Sequência Molecular , Proteínas Recombinantes/imunologia
20.
Protein Expr Purif ; 12(1): 75-84, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9473460

RESUMO

The hepatitis E virus (HEV) capsid antigen has been proposed as a candidate subunit vaccine for the prevention of hepatitis E. The full-length HEV ORF2 protein product is predicted to contain 660 amino acids and to weigh 72,000 daltons. Expression of the HEV ORF2 capsid gene from recombinant baculoviruses in insect cells produced multiple immunoreactive proteins ranging in size from 30 to 100 kDa. The most abundant HEV proteins had molecular weights of 72, 63, 56, and 53 kDa. Temporal expression kinetics of these viral polypeptides indicated that the 72- and 63-kDa polypeptides were produced abundantly within the initial 36 h. postinfection but were replaced by 56- and 53-kDa polypeptides in the cell and medium, respectively, by 48 h postinfection. The 53-kDa protein was secreted as early as 24 h. postinfection, and accumulation in the medium peaked by 72 h postinfection. Purification of the 53-, 56-, and 63-kDa viral polypeptides was accomplished by anion-exchange and subsequent gel filtration chromatography. Sequence analysis of the 53-, 56-, and 63-kDa HEV polypeptides indicated that the amino terminus was amino acid residue 112 of the predicted full-length protein product. The results of carboxy terminal amino acid sequencing indicated that the carboxy terminus of the 53-, 56-, and 63-kDa HEV proteins was located at amino acid residues 578, 607, and 660, respectively. The molecular masses of the 53- and 56-kDa HEV polypeptides were 53,872 and 56,144 as determined by mass spectroscopy.


Assuntos
Vírus da Hepatite E/química , Proteínas Virais/química , Sequência de Aminoácidos , Aminoácidos/análise , Animais , Antígenos Virais/química , Antígenos Virais/genética , Baculoviridae/genética , Sequência de Bases , Capsídeo/química , Capsídeo/genética , Capsídeo/imunologia , Células Clonais , Primers do DNA/genética , Expressão Gênica , Hepatite E/prevenção & controle , Vírus da Hepatite E/genética , Vírus da Hepatite E/imunologia , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Spodoptera , Vacinas contra Hepatite Viral/genética , Vacinas contra Hepatite Viral/isolamento & purificação , Proteínas Virais/genética , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA