Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Sci Adv ; 9(42): eadj4198, 2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37862421

RESUMO

Virus-induced changes in host lipid metabolism are an important but poorly understood aspect of viral pathogenesis. By combining nontargeted lipidomics analyses of infected cells and purified extracellular quasi-enveloped virions with high-throughput RNA sequencing and genetic depletion studies, we show that hepatitis A virus, an hepatotropic picornavirus, broadly manipulates the host cell lipid environment, enhancing synthesis of ceramides and other sphingolipids and transcriptionally activating acyl-coenzyme A synthetases and fatty acid elongases to import and activate long-chain fatty acids for entry into the fatty acid elongation cycle. Phospholipids with very-long-chain acyl tails (>C22) are essential for genome replication, whereas increases in sphingolipids support assembly and release of quasi-enveloped virions wrapped in membranes highly enriched for sphingomyelin and very-long-chain ceramides. Our data provide insight into how a pathogenic virus alters lipid flux in infected hepatocytes and demonstrate a distinction between lipid species required for viral RNA synthesis versus nonlytic quasi-enveloped virus release.


Assuntos
Hepatovirus , RNA Viral , Hepatovirus/metabolismo , RNA Viral/genética , Replicação do RNA , Liberação de Vírus , Replicação Viral/fisiologia , Ácidos Graxos/metabolismo , Esfingolipídeos , Ceramidas
2.
J Virol ; 96(21): e0119522, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-36286484

RESUMO

Hepatoviruses are atypical hepatotropic picornaviruses that are released from infected cells without lysis in small membranous vesicles. These exosome-like, quasi-enveloped virions (eHAV) are infectious and the only form of hepatitis A virus (HAV) found circulating in blood during acute infection. eHAV is released through multivesicular endosomes in a process dependent on endosomal sorting complexes required for transport (ESCRT). Capsid protein interactions with the ESCRT-associated Bro1 domain proteins, ALG-2-interacting protein X (ALIX) and His domain-containing protein tyrosine phosphatase (HD-PTP), which are both recruited to the pX domain of 1D (VP1pX), are critical for this process. Previous proteomics studies suggest pX also binds the HECT domain, NEDD4 family E3 ubiquitin ligase, ITCH. Here, we confirm this interaction and show ITCH binds directly to the carboxy-terminal half of pX from both human and bat hepatoviruses independently of ALIX. A small chemical compound (compound 5) designed to disrupt interactions between WW domains of NEDD4 ligases and substrate molecules blocked ITCH binding to pX and demonstrated substantial antiviral activity against HAV. CRISPR deletion or small interfering RNA (siRNA) knockdown of ITCH expression inhibited the release of a self-assembling nanocage protein fused to pX and also impaired the release of eHAV from infected cells. The release could be rescued by overexpression of wild-type ITCH, but not a catalytically inactive ITCH mutant. Despite this, we found no evidence that ITCH ubiquitylates pX or that eHAV release is strongly dependent upon Lys residues in pX. These data indicate ITCH plays an important role in the ESCRT-dependent release of quasi-enveloped hepatovirus, although the substrate molecule targeted for ubiquitylation remains to be determined. IMPORTANCE Mechanisms underlying the cellular release of quasi-enveloped hepatoviruses are only partially understood, yet play a crucial role in the pathogenesis of this common agent of viral hepatitis. Multiple NEDD4 family E3 ubiquitin ligases, including ITCH, have been reported to promote the budding of conventional enveloped viruses but are not known to function in the release of HAV or other picornaviruses from infected cells. Here, we show that the unique C-terminal pX extension of the VP1 capsid protein of HAV interacts directly with ITCH and that ITCH promotes eHAV release in a manner analogous to its role in budding of some conventional enveloped viruses. The catalytic activity of ITCH is required for efficient eHAV release and may potentially function to ubiquitylate the viral capsid or activate ESCRT components.


Assuntos
Vírus da Hepatite A , Ubiquitina-Proteína Ligases , Humanos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Hepatovirus/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vírus da Hepatite A/fisiologia , Ubiquitina-Proteína Ligases Nedd4/metabolismo
3.
J Virol ; 82(20): 10118-28, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684812

RESUMO

Internally located, cis-acting RNA replication elements (cre) have been identified within the genomes of viruses representing each of the major picornavirus genera (Enterovirus, Rhinovirus, Aphthovirus, and Cardiovirus) except Hepatovirus. Previous efforts to identify a stem-loop structure with cre function in hepatitis A virus (HAV), the type species of this genus, by phylogenetic analyses or thermodynamic predictions have not succeeded. However, a region of markedly suppressed synonymous codon variability was identified in alignments of HAV sequences near the 5' end of the 3D(pol)-coding sequence of HAV, consistent with noncoding constraints imposed by an underlying RNA secondary structure. Subsequent MFOLD predictions identified a 110-nucleotide (nt) complex stem-loop in this region with a typical AAACA/G cre motif in its top loop. A potentially homologous RNA structure was identified in this region of the avian encephalitis virus genome, despite little nucleotide sequence relatedness between it and HAV. Mutations that disrupted secondary RNA structure or the AAACA/G motif, without altering the amino acid sequence of 3D(pol), ablated replication of a subgenomic HAV replicon in transfected human hepatoma cells. Replication competence could be rescued by reinsertion of the native 110-nt stem-loop structure (but not an abbreviated 45-nt stem-loop) upstream of the HAV coding sequence in the replicon. These results suggest that this stem-loop is functionally similar to cre elements of other picornaviruses and likely involved in templating VPg uridylylation as in other picornaviruses, despite its significantly larger size and lower free folding energy.


Assuntos
Sequência de Bases , Hepatovirus/genética , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/genética , Sequência de Aminoácidos , Animais , Genoma Viral , Hepatovirus/metabolismo , Humanos , Dados de Sequência Molecular , Mutação , RNA Viral/metabolismo , Replicon/genética , Replicação Viral/genética
4.
J Gen Virol ; 86(Pt 10): 2747-2751, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186228

RESUMO

Although Hepatitis A virus (HAV) is transmitted by the faecal-oral route, its target for replication is the liver. Little is known of its interactions with cells of the gastrointestinal tract, and it is not known by which mechanisms HAV crosses the intestinal epithelium. In this study, it is shown that HAV associated with IgA is translocated from the apical to the basolateral compartment of polarized epithelial cells via the polymeric immunoglobulin receptor by IgA-mediated reverse transcytosis. The relevance of this mechanism, by which HAV-IgA complexes may overcome the intestinal barrier and contribute to infections of the liver, results from the fact that HAV-IgA complexes are infectious for hepatocytes and that significant amounts of intestinal HAV-IgA are present during acute infections, which are also partly transmitted. Besides supporting the primary infection, this mechanism may play a role in relapsing infections by establishing an enterohepatic cycle for HAV.


Assuntos
Células Epiteliais/virologia , Hepatovirus/metabolismo , Imunoglobulina A/imunologia , Receptores de Imunoglobulina Polimérica/fisiologia , Complexo Antígeno-Anticorpo/química , Transporte Biológico , Membrana Celular/virologia , Polaridade Celular , Hepatovirus/imunologia , Humanos , Células Tumorais Cultivadas
5.
Artigo em Russo | MEDLINE | ID: mdl-11548267

RESUMO

On the basis of coding regions on the fragments of genes P1 and P2 of hepatitis A virus (HAV) recombinant proteins of this virus have been synthesized in the prokaryotic expressing system of E. coli, isolated and studied with the use of sera obtained from hepatitis A patients. The capacity of HAV recombinant proteins for binding with the sera of patients with hepatitis A in the acute stage has been shown with the use of immunoblotting and the indirect solid-phase enzyme immunoassay. The results obtained in this investigation are discussed in the light of the possible use of recombinant proteins for the detection of HAV markers.


Assuntos
Escherichia coli/metabolismo , Genoma Viral , Hepatovirus/metabolismo , Poliproteínas/metabolismo , Proteínas Virais/metabolismo , Proteínas Sanguíneas/metabolismo , Western Blotting , Escherichia coli/genética , Hepatite A/sangue , Hepatovirus/genética , Humanos , Técnicas Imunoenzimáticas , Poliproteínas/genética , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Virais/genética
6.
J Virol ; 75(17): 7854-63, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11483729

RESUMO

The question of whether translation initiation factor eIF4E and the complete eIF4G polypeptide are required for initiation dependent on the IRES (internal ribosome entry site) of hepatitis A virus (HAV) has been examined using in vitro translation in standard and eIF4G-depleted rabbit reticulocyte lysates. In agreement with previous publications, the HAV IRES is unique among all picornavirus IRESs in that it was inhibited if translation initiation factor eIF4G was cleaved by foot-and-mouth disease L-proteases. In addition, the HAV IRES was inhibited by addition of eIF4E-binding protein 1, which binds tightly to eIF4E and sequesters it, thus preventing its association with eIF4G. The HAV IRES was also inhibited by addition of m(7)GpppG cap analogue, irrespective of whether the RNA tested was capped or not. Thus, initiation on the HAV IRES requires that eIF4E be associated with eIF4G and that the cap-binding pocket of eIF4E be empty and unoccupied. This suggests two alternative models: (i) initiation requires a direct interaction between an internal site in the IRES and eIF4E/4G, an interaction which involves the cap-binding pocket of eIF4E in addition to any direct eIF4G-RNA interactions; or (ii) it requires eIF4G in a particular conformation which can be attained only if eIF4E is bound to it, with the cap-binding pocket of the eIF4E unoccupied.


Assuntos
Regiões 5' não Traduzidas , Hepatovirus/genética , Fatores de Iniciação de Peptídeos/metabolismo , Capuzes de RNA/metabolismo , Ribossomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Proteínas de Ciclo Celular , Linhagem Celular , Fosfatos de Dinucleosídeos/farmacologia , Endopeptidases/genética , Endopeptidases/metabolismo , Fator de Iniciação 4E em Eucariotos , Fator de Iniciação Eucariótico 4G , Hepatovirus/metabolismo , Humanos , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacologia , Biossíntese de Proteínas , Análogos de Capuz de RNA , RNA Mensageiro/metabolismo , Coelhos , Proteínas Recombinantes , Transcrição Gênica
7.
J Virol ; 75(17): 7864-71, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11483730

RESUMO

The hepatitis A virus (HAV) internal ribosome entry segment (IRES) is unique among the picornavirus IRESs in that it is inactive in the presence of either the entero- and rhinovirus 2A or aphthovirus Lb proteinases. Since these proteinases both cleave eukaryotic initiation factor 4G (eIF4G) and HAV IRES activity could be rescued in vitro by addition of eIF4F to proteinase-treated extracts, it was concluded that the HAV IRES requires eIF4F containing intact eIF4G. Here, we show that the inability of the HAV IRES to function with cleaved eIF4G cannot be attributed to inefficient binding of the cleaved form of eIF4G by the HAV IRES. Indeed, the binding of both intact eIF4F and the C-terminal cleavage product of eIF4G to the HAV IRES was virtually indistinguishable from their binding to the encephalomyocarditis virus IRES, as assessed by UV cross-linking and filter retention assays. Rather, we show that HAV IRES activity requires, either directly or indirectly, components of the eIF4F complex which interact with the N-terminal fragment of eIF4G. Effectively, HAV IRES activity, but not that of the human rhinovirus IRES, was sensitive to the rotavirus nonstructural protein NSP3 [which displaces poly(A)-binding protein from the eIF4F complex], to recombinant eIF4E-binding protein (which prevents the association of the cap binding protein eIF4E with eIF4G), and to cap analogue.


Assuntos
Regiões 5' não Traduzidas , Hepatovirus/genética , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas , Ribossomos/metabolismo , Animais , Endopeptidases/metabolismo , Fator de Iniciação 4F em Eucariotos , Fator de Iniciação Eucariótico 4G , Hepatovirus/metabolismo , Humanos , Plasmídeos/genética , Proteínas de Ligação a Poli(A) , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Coelhos , Ribossomos/genética , Transcrição Gênica
8.
J Virol ; 75(2): 717-25, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134285

RESUMO

Hepatitis A virus (HAV) infects African green monkey kidney (AGMK) cells via the HAV cellular receptor-1 (havcr-1), a mucin-like type 1 integral-membrane glycoprotein of unknown natural function. The ectodomain of havcr-1 contains an N-terminal immunoglobulin-like cysteine-rich region (D1), which binds protective monoclonal antibody (MAb) 190/4, followed by an O-glycosylated mucin-like threonine-serine-proline-rich region that extends D1 well above the cell surface. To study the interaction of HAV with havcr-1, we constructed immunoadhesins fusing the hinge and Fc portion of human IgG1 to D1 (D1-Fc) or the ectodomain of the poliovirus receptor (PVR-Fc) and expressed them in CHO cells. These immunoadhesins were secreted to the cell culture medium and purified through protein A-agarose columns. In a solid-phase assay, HAV bound to D1-Fc in a concentration-dependent manner whereas background levels of HAV bound to PVR-Fc. Binding of HAV to D1-Fc was blocked by treatment with MAb 190/4 but not with control MAb M2, which binds to a tag epitope introduced between the D1 and Fc portions of the immunoadhesin. D1-Fc neutralized approximately 1 log unit of the HAV infectivity in AGMK cells, whereas PVR-Fc had no effect in the HAV titers. A similarly poor reduction in HAV titers was observed after treating the same stock of HAV with murine neutralizing MAbs K2-4F2, K3-4C8, and VHA 813. Neutralization of poliovirus by PVR-Fc but not by D1-Fc indicated that the virus-receptor interactions were specific. These results show that D1 is sufficient for binding and neutralization of HAV and provide further evidence that havcr-1 is a functional cellular receptor for HAV.


Assuntos
Hepatovirus/metabolismo , Imunoglobulinas/química , Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/química , Receptores Virais/química , Animais , Anticorpos Monoclonais/metabolismo , Células CHO , Cricetinae , Cisteína , Epitopos , Receptor Celular 1 do Vírus da Hepatite A , Hepatovirus/imunologia , Hepatovirus/fisiologia , Humanos , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Testes de Neutralização , Complexo de Proteínas do Centro de Reação Fotossintética , Complexo de Proteína do Fotossistema II , Receptores Fc/metabolismo , Receptores Virais/imunologia , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
9.
J Virol ; 74(14): 6459-68, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10864658

RESUMO

Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), a cellular enzyme involved in glycolysis, binds specifically to several viral RNAs, but the functional significance of this interaction is uncertain. Both GAPDH and polypyrimidine tract binding protein (PTB) bind to overlapping sites in stem-loop IIIa of the internal ribosome entry site (IRES) of Hepatitis A virus (HAV), a picornavirus. Since the binding of GAPDH destabilizes the RNA secondary structure, we reasoned that GAPDH may suppress the ability of the IRES to direct cap-independent translation, making its effects antagonistic to the translation-enhancing activity of PTB (D. E. Schultz, C. C. Hardin, and S. M. Lemon, J. Biol. Chem. 271:14134-14142, 1996). To test this hypothesis, we constructed plasmids containing a dicistronic transcriptional unit in which the HAV IRES was placed between an upstream GAPDH-coding sequence and a downstream Renilla luciferase (RLuc) sequence. Transfection with this plasmid results in overexpression of GAPDH and in RLuc production as a measure of IRES activity. RLuc activity was compared with that from a control, null-expression plasmid that was identical except for a frameshift mutation within the 5' GAPDH coding sequence. In transfection experiments, GAPDH overexpression significantly suppressed HAV IRES activity in BSC-1 and FRhK-4 cells but not in Huh-7 cells, which have a significantly greater cytoplasmic abundance of PTB. GAPDH suppression of HAV translation was greater with the wild-type HAV IRES than with the IRES from a cell culture-adapted virus (HM175/P16) that has reproducibly higher basal translational activity in BSC-1 cells. Stem-loop IIIa RNA from the latter IRES had significantly lower affinity for GAPDH in filter binding experiments. Thus, the binding of GAPDH to the IRES of HAV suppresses cap-independent viral translation in vivo in African green monkey kidney cells. The enhanced replication capacity of cell culture-adapted HAV in such cells may be due in part to reduced affinity of the viral IRES for GAPDH.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Hepatovirus/fisiologia , Fragmentos de Peptídeos/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribossomos/fisiologia , Linhagem Celular , Células HeLa , Hepatovirus/metabolismo , Humanos , Immunoblotting , Luciferases/biossíntese , Luciferases/genética , Mutação , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Ligação Proteica , Biossíntese de Proteínas , Ribossomos/genética , Ribossomos/metabolismo , Transfecção , Replicação Viral
10.
J Gen Virol ; 81(Pt 5): 1373-81, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10769081

RESUMO

The viral protein 2A of hepatitis A virus (HAV) lacks the conserved 18 aa sequence found in other picornavirus proteases; hence, it is unclear whether the induction of CPE by culture-adapted HAV strains is due to 2A-mediated activity. Moreover, the cleavage sites and actual borders of HAV 2A are not known. Accordingly, a nested series of cDNA sequences encoding the segment of the HAV polyprotein (aa 760-1087) were linked to the 5'-UTR of poliovirus type 2 (Lansing strain) and inserted downstream of the gene encoding human growth hormone (GH). Following transfection of COS-1 cells, levels of GH (translation of which was entirely cap dependent) were determined in culture supernatants. Expression of HAV peptides extending from aa 764, 776 or 791 to 981 strongly inhibited cap-dependent translation of GH, whereas cap-independent expression of a reporter gene (CAT) directed by the poliovirus RNA 5'-UTR was unaffected. The inhibitory effect was absent in constructs expressing either the short peptide encompassing aa 760-836 or proteins initiated downstream of the putative cleavage site 836-837, suggesting that the boundaries of a functional HAV 2A may extend from the Gln/Ser junction 791-792 to residue 981, while peptides initiated at the Gln/Ala pair 836-837 may result from alternative cleavage. Point mutations that substituted members of the triad Ser(916), His(927) and Asp(931) abolished the inhibitory effect on cap-dependent translation, suggesting that the HAV-induced CPE may be mediated by 2A protein.


Assuntos
Cisteína Endopeptidases/genética , Cisteína Endopeptidases/fisiologia , Expressão Gênica , Hepatovirus/metabolismo , Capuzes de RNA , Proteínas Virais , Regiões 5' não Traduzidas , Animais , Células COS , Cisteína Endopeptidases/metabolismo , Efeito Citopatogênico Viral , Hepatovirus/genética , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Plasmídeos , Reação em Cadeia da Polimerase , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Transfecção
11.
Virology ; 264(2): 410-21, 1999 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-10562502

RESUMO

The small hydrophobic protein 3AB of the picornaviruses, encompassing the replication primer 3B, has been suggested to anchor the viral replication complex to membranes. For hepatitis A virus (HAV) 3AB, we have previously demonstrated its ability to form stable homodimers, to bind to membranes, and to interact specifically with RNA, implicating its multiple involvement in viral replication. In the present report, we show that HAV 3AB additionally interacts with HAV protein 3CD, a feature also described for the corresponding polypeptide of poliovirus. By assessing the interactions of three deletion mutants, distinct domains of HAV 3AB were mapped. The hydrophobic domain and the 3B moiety were found to be essential for the 3AB interaction with 3CD. Both electrostatic and hydrophobic forces are involved in this interaction. The cluster of charged amino acid residues at the C terminus of 3A seems to determine the specificity of 3AB interaction with RNA structures formed at either terminus of the HAV genome. Furthermore, our data implicate that 3A can interact with HAV RNA. Compared with poliovirus 3AB, which by itself is a nonspecific RNA-binding protein, HAV 3AB specifically recognizes HAV RNA structures that might be of relevance for initiation of viral RNA replication.


Assuntos
Cisteína Endopeptidases/metabolismo , Hepatovirus/fisiologia , RNA Viral/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas Virais , Proteases Virais 3C , Sequência de Aminoácidos , Sítios de Ligação , Mapeamento Cromossômico , Cisteína Endopeptidases/genética , Dimerização , Hepatovirus/genética , Hepatovirus/metabolismo , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Proteínas do Core Viral/genética
12.
J Virol ; 73(12): 9867-78, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559299

RESUMO

The orchestrated liberation of viral proteins by 3C(pro)-mediated proteolysis is pivotal for gene expression by picornaviruses. Proteolytic processing is regulated either by the amino acid sequence at the cleavage site of the substrate or by cofactors covalently or noncovalently linked to the viral proteinase. To determine the role of the amino acid sequence at cleavage sites 3A/3B and 3B/3C that are essential for the liberation of 3C(pro) from its precursors and to assess the function of the stable processing intermediates 3AB and 3ABC, we studied the effect of cleavage site mutations on hepatitis A virus (HAV) polyprotein processing, particle formation, and replication. Using the recombinant vaccinia virus system, we showed that the normally retarded cleavage at the 3A/3B junction can be improved by altering the amino acid sequence at the scissile bond such that it matches the preferred HAV 3C cleavage sites. In contrast to the processing products of the wild-type polyprotein, 3ABC was no longer detectable in the mutant. VP0 and VP3 were generated less efficiently, implying that processing of the structural protein precursor P1-2A depends on the presence of stable 3ABC and/or 3AB. In addition, cleavage of 2BC was impaired in 3AB/3ABC-deficient mutants. Formation of HAV particles was not affected in mutants with blocked 3A/3B and/or 3B/3C cleavage sites. However, 3ABC-deficient mutants produced small numbers of HAV particles, which could be augmented by coexpressing 3AB or 3ABC. The hydrophobic domain of 3A that has been proposed to mediate membrane anchorage of the replication complex was crucial for restoration of defective particle formation. In vitro transcripts of the various cleavage site mutants were unable to initiate an infectious cycle, and no progeny viruses were obtained even after blind passages. Taken together, the data suggest that accumulation of uncleaved HAV 3AB and/or 3ABC is pivotal for both viral replication and efficient particle formation.


Assuntos
Hepatovirus/metabolismo , Poliproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Montagem de Vírus , Animais , Sítios de Ligação , Células COS , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Endopeptidases , Hepatovirus/genética , Mutagênese , Poliproteínas/genética , Proteínas do Core Viral/genética , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Vírion , Replicação Viral
13.
J Virol ; 73(8): 6220-7, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400711

RESUMO

Most details of the processing of the hepatitis A virus (HAV) polyprotein are known. Unique among members of the family Picornaviridae, the primary cleavage of the HAV polyprotein is mediated by 3Cpro, the only proteinase known to be encoded by the virus, at the 2A/2B junction. All other cleavages of the polyprotein have been considered to be due to 3Cpro, although the precise location and mechanism responsible for the VP1/2A cleavage have been controversial. Here we present data that argue strongly against the involvement of the HAV 3Cpro proteinase in the maturation of VP1 from its VP1-2A precursor. Using a heterologous expression system based on recombinant vaccinia viruses directing the expression of full-length or truncated capsid protein precursors, we show that the C terminus of the mature VP1 capsid protein is located near residue 764 of the polyprotein. However, a proteolytically active HAV 3Cpro that was capable of directing both VP0/VP3 and VP3/VP1 cleavages in vaccinia virus-infected cells failed to process the VP1-2A precursor. Using site-directed mutagenesis of an infectious molecular clone of HAV, we modified potential VP1/2A cleavage sites that fit known 3Cpro recognition criteria and found that a substitution that ablates the presumed 3Cpro dipeptide recognition sequence at Glu764-Ser765 abolished neither infectivity nor normal VP1 maturation. Altered electrophoretic mobility of VP1 from a viable mutant virus with an Arg764 substitution indicated that this residue is present in VP1 and that the VP1/2A cleavage occurs downstream of this residue. These data indicate that maturation of the HAV VP1 capsid protein is not dependent on 3Cpro processing and may thus be uniquely dependent on a cellular proteinase.


Assuntos
Cisteína Endopeptidases/metabolismo , Hepatovirus/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais , Proteínas Estruturais Virais/metabolismo , Proteases Virais 3C , Substituição de Aminoácidos , Animais , Linhagem Celular , Cisteína Endopeptidases/genética , Vetores Genéticos , Ácido Glutâmico , Humanos , Macaca mulatta , Mutagênese Sítio-Dirigida , Peptídeos/genética , Peptídeos/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Recombinação Genética , Vaccinia virus , Proteínas Estruturais Virais/genética , Replicação Viral
14.
J Biol Chem ; 274(8): 4527-31, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-9988685

RESUMO

Capsid assembly is the final event of virus replication, and its understanding is pivotal for the design of empty capsid-based recombinant vaccines and drug delivery systems. Although the capsid structure of several members of the picornavirus family has been elucidated, little is known about the structural elements governing the assembly process that is tightly associated with proteolytic processing of the viral polyprotein. Among the picornaviruses, hepatitis A virus (HAV) is unique in that it contains VP1-2A as a structural component and the small structural protein VP4, which argues for an assembly pathway different from that proposed for other picornaviruses. Using a recombinant system we show here that proteolytic processing of the HAV capsid proteins' precursor P1-2A is independent of the terminal domains 2A and VP4 of the substrate. However, both terminal domains play distinct roles in the assembly of viral particles. 2A as part of P1-2A is a primary signal for the assembly of pentameric structures which only further aggregate to empty viral capsids when VP4 is present as the N terminus of the precursor. Particle formation in the hepatovirus genus is thus regulated by two intrinsic signals that are distinct from those described for other picornaviruses.


Assuntos
Proteínas do Capsídeo , Capsídeo/metabolismo , Cisteína Endopeptidases/metabolismo , Hepatovirus/metabolismo , Sinais Direcionadores de Proteínas/metabolismo , Proteínas Virais , Vírion/metabolismo , Animais , Sequência de Bases , Células COS , Primers do DNA , Hidrólise
15.
Acta Virol ; 43(5): 291-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10757229

RESUMO

Cleavage of VP0 to VP2 via intramolecular scission is known as the viral maturation cleavage, as VP0 is found in immature particles, whilst VP2 is found in mature particles. The effect of low pH on the kinetics of hepatitis A virus (HAV) capsid protein VP0 cleavage in provirions was examined by Western blot analysis. VP0 scission was found to be dramatically enhanced under acidic conditions, similar to those encountered on entry of virus particles into the cell via endocytosis. The cleavage of VP0 to VP2 led to an increase in the specific infectivity of viral particles, indicating that mature virions are more infectious than immature provirions. The data are consistent with a model where conformational changes induced by low pH aid scission of VP0, and the increase in kinetics of VP0 cleavage may have relevance for viral uncoating, as only mature HAV particles are thought capable of uncoating within the host cell.


Assuntos
Capsídeo/metabolismo , Hepatovirus/metabolismo , Animais , Western Blotting , Proteínas do Capsídeo , Linhagem Celular , Chlorocebus aethiops , Hepatovirus/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Cinética , Provírus/metabolismo , Vírion/fisiologia
16.
J Virol ; 72(12): 9668-75, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9811700

RESUMO

Utilization of internal ribosome entry segment (IRES) structures in the 5' noncoding region (5'NCR) of picornavirus RNAs for initiation of translation requires a number of host cell factors whose distribution may vary in different cells and whose requirement may vary for different picornaviruses. We have examined the requirement of the cellular protein poly(rC) binding protein 2 (PCBP2) for hepatitis A virus (HAV) RNA translation. PCBP2 has recently been identified as a factor required for translation and replication of poliovirus (PV) RNA. PCBP2 was shown to be present in FRhK-4 cells, which are permissive for growth of HAV, as it is in HeLa cells, which support translation of HAV RNA but which have not been reported to host replication of the virus. Competition RNA mobility shift assays showed that the 5'NCR of HAV RNA competed for binding of PCBP2 with a probe representing stem-loop IV of the PV 5'NCR. The binding site on HAV RNA was mapped to nucleotides 1 to 157, which includes a pyrimidine-rich sequence. HeLa cell extracts that had been depleted of PCBP2 by passage over a PV stem-loop IV RNA affinity column supported only low levels of HAV RNA translation. Translation activity was restored upon addition of recombinant PCBP2 to the depleted extract. Removal of the 5'-terminal 138 nucleotides of the HAV RNA, or removal of the entire IRES, eliminated the dependence of HAV RNA translation on PCBP2.


Assuntos
Proteínas de Ligação a DNA , Hepatovirus/genética , Hepatovirus/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas , Biossíntese de Proteínas , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição , Sítios de Ligação/genética , Ligação Competitiva , Linhagem Celular , Células HeLa , Hepatovirus/crescimento & desenvolvimento , Humanos , Picornaviridae/genética , Picornaviridae/metabolismo , Ribossomos/genética , Ribossomos/metabolismo
17.
Biochem Biophys Res Commun ; 249(1): 266-74, 1998 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-9705870

RESUMO

Hepatitis A virus (HAV) protein 3AB is a membrane-interacting protein containing a stretch of 21 hydrophobic amino acid residues. The nature of its membrane association was studied in detail by analysing various deletion mutants. In vivo and in vitro expression of the wild-type protein and its mutants allowed to demonstrate that the hydrophobic domain interacts with membranes and to define the portions essential for this feature. Furthermore, the results suggest that 3AB behaves as an integral membrane protein. Expression in Escherichia coli showed that 3AB can be isolated, in association with membranes, both in monomeric and in dimeric form. This finding was confirmed in vitro after post-translational incubation of the protein with microsomal membranes. Analysis of deletion mutants demonstrated that the dimerization region colocalises with the hydrophobic transmembrane domain, implicating that HAV 3AB could form oligomers mediated by the interaction of transmembrane alpha-helices.


Assuntos
Hepatovirus/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Core Viral/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Dimerização , Escherichia coli , Humanos , Proteínas de Membrana/genética , Microssomos/metabolismo , Dados de Sequência Molecular , Mutação , Proteínas do Core Viral/genética
18.
Biotechnol Appl Biochem ; 28(1): 85-94, 1998 08.
Artigo em Inglês | MEDLINE | ID: mdl-9693093

RESUMO

CSL Limited, an Australian biopharmaceutical company, has recently converted its method of manufacture for human albumin from a traditional Cohn-ethanol fractionation method to a method employing chromatographic techniques. Studies were undertaken to determine the efficiency of the chromatographic and pasteurization steps used in the manufacture of Albumex(R) (CSL's trade name for albumin) in removing and inactivating the potential viral contaminant, hepatitis A virus (HAV). The manufacturing process for Albumex(R) includes three chromatographic steps, two of which are ion-exchange steps (DEAE-Sepharose(R) Fast Flow and CM-Sepharose(R) Fast Flow) and the third is a gel-filtration step (Sephacryl(R) S200 HR). The final stage of the Albumex(R) process involves a bulk pasteurization step where product is held at 60 degrees C for 10 h. HAV partitioning experiments on the DEAE-Sepharose(R) FF and CM-Sepharose(R) FF ion-exchange and Sephacryl(R) S200 HR gel-filtration columns were performed with scaled-down models of the production-scale chromatographic Albumex(R) process. Production samples collected before each of the chromatographic steps were spiked with HAV and processed through each of the scaled-down chromatographic columns. Samples collected during processing were assayed and the log10 reduction factors calculated. Inactivation kinetics of HAV were examined during the pasteurization of Albumex(R) 5 and 20 [5% and 20% (w/v) albumin solutions] held at 60 degrees C for 10 h. Log10 reductions for HAV through the DEAE-Sepharose(R) FF, CM-Sepharose(R) FF and Sephacryl(R) S200 HR chromatographic columns were 5.3, 1.5 and 4.2 respectively, whereas a 4.4 and a greater than 3.9 log10 reduction in HAV in Albumex(R) 5 and 20 respectively were achieved during pasteurization.


Assuntos
Hepatovirus/metabolismo , Preparações Farmacêuticas/isolamento & purificação , Albumina Sérica/isolamento & purificação , Cromatografia em Gel , Cromatografia por Troca Iônica/métodos , Temperatura Alta , Humanos , Albumina Sérica/uso terapêutico , Temperatura
19.
J Virol ; 72(8): 6621-8, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9658108

RESUMO

The hepatitis A virus cellular receptor 1 (HAVcr-1) cDNA was isolated from a cDNA expression library of African green monkey kidney (AGMK) cells by using protective monoclonal antibody (MAb) 190/4, which blocks the binding of hepatitis A virus (HAV) to AGMK cells. The HAVcr-1 cDNA codes for havcr-1, a 451-amino-acid class I integral-membrane mucin-like glycoprotein of unknown natural function. To determine the existence of a human homolog(s) of HAVcr-1 (huHAVcr-1), we used HAVcr-1-specific primers to amplify cDNAs from human liver and kidney mRNA by reverse transcription-PCR. Nucleotide sequence analysis revealed that the amplified liver and kidney huHAVcr-1 cDNAs were identical and that they coded for a 359-amino-acid glycoprotein, termed huhavcr-1, which was approximately 79% identical to havcr-1. The six Cys residues of the extracellular domain of havcr-1 and its first N-glycosylation site were conserved in huhavcr-1. However, the number of hexameric repeats of the mucin-like region was reduced from 27 in havcr-1 to 13 in huhavcr-1. In addition, 12 C-terminal amino acids in the cytoplasmic domain of huhavcr-1 were deleted. Northern blot analysis of poly(A) RNA showed that huhavcr-1 is expressed in every organ analyzed, including the liver, small intestine, colon, and spleen, and that it is expressed at higher levels in the kidney and testis. Although dog cells transfected with the huHAVcr-1 cDNA did not express the protective 190/4 epitope, they bound hepatitis A virus (HAV) and gained limited susceptibility to HAV infection. Treatment with MAb 190/4 did not protect AGMK cell transfectants expressing huhavcr-1 against HAV, suggesting that HAV infected these cells via the huhavcr-1 receptor and not the endogenously expressed havcr-1, which was blocked by MAb 190/4. Our data demonstrate that huhavcr-1 is a binding receptor for HAV and suggest that it is also a functional receptor for HAV.


Assuntos
Hepatovirus/metabolismo , Glicoproteínas de Membrana/genética , Receptores Virais/genética , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Sequência de Bases , Linhagem Celular , Clonagem Molecular , DNA Complementar , Cães , Receptor Celular 1 do Vírus da Hepatite A , Hepatovirus/fisiologia , Humanos , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Coelhos , Receptores Virais/imunologia , Receptores Virais/metabolismo , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção
20.
Arch Virol ; 143(5): 931-44, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9645199

RESUMO

The direct function of hepatitis A virus (HAV) protein 2C, a putative NTPase, is not known, yet genetic evidence obtained from chimeric viruses carrying the 2C genomic region of different HAV variants indicates that it plays a pivotal role in viral replication. In a first assessment of its potential function(s), membrane and RNA binding properties of HAV 2C were studied after expressing the protein in various recombinant systems. In contrast to poliovirus 2C, expression of HAV 2C was inhibitory to the growth and protein synthesis of bacteria. Deletion of the N-terminal amphipathic helix of 2C abrogated this effect and the ability of 2C to associate with eukaryotic membranes. Both, purified 2C and the N-terminally truncated protein were shown to bind RNA in vitro. Our data taken together suggest that HAV 2C is a multifunctional protein.


Assuntos
Hepatovirus/metabolismo , RNA/metabolismo , Proteínas Virais/metabolismo , Animais , Células COS , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Escherichia coli/genética , Expressão Gênica , Genes Virais , Hepatovirus/genética , Membranas/metabolismo , Poliovirus/genética , Poliovirus/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...